Network


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

Hotspot


Dive into the research topics where Lixia Diao is active.

Publication


Featured researches published by Lixia Diao.


Clinical Cancer Research | 2013

An Epithelial–Mesenchymal Transition Gene Signature Predicts Resistance to EGFR and PI3K Inhibitors and Identifies Axl as a Therapeutic Target for Overcoming EGFR Inhibitor Resistance

Lauren Averett Byers; Lixia Diao; Jing Wang; Pierre Saintigny; Luc Girard; Michael Peyton; Li Shen; Youhong Fan; Uma Giri; Praveen Tumula; Monique B. Nilsson; Jayanthi Gudikote; Hai T. Tran; Robert J. Cardnell; David J. Bearss; Steven L. Warner; Jason M. Foulks; Steven B. Kanner; Varsha Gandhi; Nancy L. Krett; Steven T. Rosen; Edward S. Kim; Roy S. Herbst; George R. Blumenschein; J. Jack Lee; Scott M. Lippman; K. Kian Ang; Gordon B. Mills; Waun Ki Hong; John N. Weinstein

Purpose: Epithelial–mesenchymal transition (EMT) has been associated with metastatic spread and EGF receptor (EGFR) inhibitor resistance. We developed and validated a robust 76-gene EMT signature using gene expression profiles from four platforms using non–small cell lung carcinoma (NSCLC) cell lines and patients treated in the Biomarker-Integrated Approaches of Targeted Therapy for Lung Cancer Elimination (BATTLE) study. Experimental Design: We conducted an integrated gene expression, proteomic, and drug response analysis using cell lines and tumors from patients with NSCLC. A 76-gene EMT signature was developed and validated using gene expression profiles from four microarray platforms of NSCLC cell lines and patients treated in the BATTLE study, and potential therapeutic targets associated with EMT were identified. Results: Compared with epithelial cells, mesenchymal cells showed significantly greater resistance to EGFR and PI3K/Akt pathway inhibitors, independent of EGFR mutation status, but more sensitivity to certain chemotherapies. Mesenchymal cells also expressed increased levels of the receptor tyrosine kinase Axl and showed a trend toward greater sensitivity to the Axl inhibitor SGI-7079, whereas the combination of SGI-7079 with erlotinib reversed erlotinib resistance in mesenchymal lines expressing Axl and in a xenograft model of mesenchymal NSCLC. In patients with NSCLC, the EMT signature predicted 8-week disease control in patients receiving erlotinib but not other therapies. Conclusion: We have developed a robust EMT signature that predicts resistance to EGFR and PI3K/Akt inhibitors, highlights different patterns of drug responsiveness for epithelial and mesenchymal cells, and identifies Axl as a potential therapeutic target for overcoming EGFR inhibitor resistance associated with the mesenchymal phenotype. Clin Cancer Res; 19(1); 279–90. ©2012 AACR.


Nature Communications | 2014

Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression

Limo Chen; Don L. Gibbons; Sangeeta Goswami; Maria Angelica Abdalla Cortez; Young Ho Ahn; Lauren Averett Byers; Xuejun Zhang; Xiaohui Yi; David Dwyer; Wei Lin; Lixia Diao; Jing Wang; Jonathon D. Roybal; Mayuri Patel; Christin Ungewiss; David H. Peng; Scott Antonia; Melanie Mediavilla-Varela; Gordon Robertson; Steve Jones; Milind Suraokar; James Welsh; Baruch Erez; Ignacio I. Wistuba; Lieping Chen; Di Peng; Shanshan Wang; Stephen E. Ullrich; John V. Heymach; Jonathan M. Kurie

Immunosuppression of tumor-infiltrating lymphocytes (TIL) is a common feature of advanced cancer, but its biological basis has remained obscure. We demonstrate here a molecular link between epithelial-to-mesenchymal transition (EMT) and CD8+ TIL immunosuppression, two key drivers of cancer progression. We show that microRNA-200 (miR-200), a cell-autonomous suppressor of EMT and metastasis, targets PD-L1. Moreover, ZEB1, an EMT activator and transcriptional repressor of miR-200, relieves miR-200 repression of PD-L1 on tumor cells, leading to CD8+ T cell immunosuppression and metastasis. These findings are supported by robust correlations between the EMT score, miR-200 levels and PD-L1 expression in multiple human lung cancer datasets. In addition to revealing a link between EMT and T cell dysfunction, these findings also show that ZEB1 promotes metastasis through a heretofore unappreciated cell non-autonomous mechanism, and suggest that subgroups of patients in whom malignant progression is driven by EMT activators may respond to treatment with PD-L1 antagonists.


Journal of the National Cancer Institute | 2012

Effect of KRAS Oncogene Substitutions on Protein Behavior: Implications for Signaling and Clinical Outcome

Nathan T. Ihle; Lauren Averett Byers; Edward S. Kim; Pierre Saintigny; Jiun-Kae Jack Lee; George R. Blumenschein; Anne Tsao; Suyu Liu; Jill E. Larsen; Jing Wang; Lixia Diao; Kevin Coombes; Lu Chen; Shuxing Zhang; Mena Abdelmelek; Ximing Tang; Vassiliki Papadimitrakopoulou; John D. Minna; Scott M. Lippman; Waun Ki Hong; Roy S. Herbst; Ignacio I. Wistuba; John V. Heymach; Garth Powis

BACKGROUND Mutations in the v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) play a critical role in cancer cell growth and resistance to therapy. Most mutations occur at codons 12 and 13. In colorectal cancer, the presence of any mutant KRas amino acid substitution is a negative predictor of patient response to targeted therapy. However, in non-small cell lung cancer (NSCLC), the evidence that KRAS mutation is a predictive factor is conflicting. METHODS We used data from a molecularly targeted clinical trial for 215 patients with tissues available out of 268 evaluable patients with refractory NSCLC to examine associations between specific mutant KRas proteins and progression-free survival and tumor gene expression. Transcriptome microarray studies of patient tumor samples and reverse-phase protein array studies of a panel of 67 NSCLC cell lines with known substitutions in KRas and in immortalized human bronchial epithelial cells stably expressing different mutant KRas proteins were used to investigate signaling pathway activation. Molecular modeling was used to study the conformations of wild-type and mutant KRas proteins. Kaplan-Meier curves and Cox regression were used to analyze survival data. All statistical tests were two-sided. RESULTS Patients whose tumors had either mutant KRas-Gly12Cys or mutant KRas-Gly12Val had worse progression-free survival compared with patients whose tumors had other mutant KRas proteins or wild-type KRas (P = .046, median survival = 1.84 months) compared with all other mutant KRas (median survival = 3.35 months) or wild-type KRas (median survival = 1.95 months). NSCLC cell lines with mutant KRas-Gly12Asp had activated phosphatidylinositol 3-kinase (PI-3-K) and mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK) signaling, whereas those with mutant KRas-Gly12Cys or mutant KRas-Gly12Val had activated Ral signaling and decreased growth factor-dependent Akt activation. Molecular modeling studies showed that different conformations imposed by mutant KRas may lead to altered association with downstream signaling transducers. CONCLUSIONS Not all mutant KRas proteins affect patient survival or downstream signaling in a similar way. The heterogeneous behavior of mutant KRas proteins implies that therapeutic interventions may need to take into account the specific mutant KRas expressed by the tumor.


Cancer Discovery | 2012

Proteomic Profiling Identifies Dysregulated Pathways in Small Cell Lung Cancer and Novel Therapeutic Targets Including PARP1

Lauren Averett Byers; Jing Wang; Monique B. Nilsson; Junya Fujimoto; Pierre Saintigny; John S. Yordy; Uma Giri; Michael Peyton; You Hong Fan; Lixia Diao; Fatemeh Masrorpour; Li Shen; Wenbin Liu; Boris Duchemann; Praveen Tumula; Vikas Bhardwaj; James Welsh; Stephanie Weber; Bonnie S. Glisson; Neda Kalhor; Ignacio I. Wistuba; Luc Girard; Scott M. Lippman; Gordon B. Mills; Kevin R. Coombes; John N. Weinstein; John D. Minna; John V. Heymach

UNLABELLED Small cell lung cancer (SCLC) is an aggressive malignancy distinct from non-small cell lung cancer (NSCLC) in its metastatic potential and treatment response. Using an integrative proteomic and transcriptomic analysis, we investigated molecular differences contributing to the distinct clinical behavior of SCLCs and NSCLCs. SCLCs showed lower levels of several receptor tyrosine kinases and decreased activation of phosphoinositide 3-kinase (PI3K) and Ras/mitogen-activated protein (MAP)/extracellular signal-regulated kinase (ERK) kinase (MEK) pathways but significantly increased levels of E2F1-regulated factors including enhancer of zeste homolog 2 (EZH2), thymidylate synthase, apoptosis mediators, and DNA repair proteins. In addition, PARP1, a DNA repair protein and E2F1 co-activator, was highly expressed at the mRNA and protein levels in SCLCs. SCLC growth was inhibited by PARP1 and EZH2 knockdown. Furthermore, SCLC was significantly more sensitive to PARP inhibitors than were NSCLCs, and PARP inhibition downregulated key components of the DNA repair machinery and enhanced the efficacy of chemotherapy. SIGNIFICANCE SCLC is a highly lethal cancer with a 5-year survival rate of less than 10%. To date, no molecularly targeted agents have prolonged survival in patients with SCLCs. As a step toward identifying new targets, we systematically profiled SCLCs with a focus on therapeutically relevant signaling pathways. Our data reveal fundamental differences in the patterns of pathway activation in SCLCs and NSCLCs and identify several potential therapeutic targets for SCLCs, including PARP1 and EZH2. On the basis of these results, clinical studies evaluating PARP and EZH2 inhibition, together with chemotherapy or other agents, warrant further investigation.


Nature Communications | 2014

A pan-cancer proteomic perspective on The Cancer Genome Atlas

Rehan Akbani; Patrick Kwok Shing Ng; Henrica Maria Johanna Werner; Maria Shahmoradgoli; Fan Zhang; Zhenlin Ju; Wenbin Liu; Ji Yeon Yang; Kosuke Yoshihara; Jun Li; Shiyun Ling; Elena G. Seviour; Prahlad T. Ram; John D. Minna; Lixia Diao; Pan Tong; John V. Heymach; Steven M. Hill; Frank Dondelinger; Nicolas Städler; Lauren Averett Byers; Funda Meric-Bernstam; John N. Weinstein; Bradley M. Broom; Roeland Verhaak; Han Liang; Sach Mukherjee; Yiling Lu; Gordon B. Mills

Protein levels and function are poorly predicted by genomic and transcriptomic analysis of patient tumors. Therefore, direct study of the functional proteome has the potential to provide a wealth of information that complements and extends genomic, epigenomic and transcriptomic analysis in The Cancer Genome Atlas (TCGA) projects. Here we use reverse-phase protein arrays to analyze 3,467 patient samples from 11 TCGA “Pan-Cancer” diseases, using 181 high-quality antibodies that target 128 total proteins and 53 post-translationally modified proteins. The resultant proteomic data is integrated with genomic and transcriptomic analyses of the same samples to identify commonalities, differences, emergent pathways and network biology within and across tumor lineages. In addition, tissue-specific signals are reduced computationally to enhance biomarker and target discovery spanning multiple tumor lineages. This integrative analysis, with an emphasis on pathways and potentially actionable proteins, provides a framework for determining the prognostic, predictive and therapeutic relevance of the functional proteome.


Cancer Discovery | 2015

Co-occurring genomic alterations define major subsets of KRAS - mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities

Ferdinandos Skoulidis; Lauren Averett Byers; Lixia Diao; Vassiliki Papadimitrakopoulou; Pan Tong; Julie Izzo; Carmen Behrens; Humam Kadara; Edwin R. Parra; Jaime Rodriguez Canales; Jianjun Zhang; Uma Giri; Jayanthi Gudikote; Maria Angelica Cortez; Chao Yang; You Hong Fan; Michael Peyton; Luc Girard; Kevin R. Coombes; Carlo Toniatti; Timothy P. Heffernan; Murim Choi; Garrett Michael Frampton; Vincent A. Miller; John N. Weinstein; Roy S. Herbst; Kwok-Kin Wong; Jianhua Zhang; Padmanee Sharma; Gordon B. Mills

UNLABELLED The molecular underpinnings that drive the heterogeneity of KRAS-mutant lung adenocarcinoma are poorly characterized. We performed an integrative analysis of genomic, transcriptomic, and proteomic data from early-stage and chemorefractory lung adenocarcinoma and identified three robust subsets of KRAS-mutant lung adenocarcinoma dominated, respectively, by co-occurring genetic events in STK11/LKB1 (the KL subgroup), TP53 (KP), and CDKN2A/B inactivation coupled with low expression of the NKX2-1 (TTF1) transcription factor (KC). We further revealed biologically and therapeutically relevant differences between the subgroups. KC tumors frequently exhibited mucinous histology and suppressed mTORC1 signaling. KL tumors had high rates of KEAP1 mutational inactivation and expressed lower levels of immune markers, including PD-L1. KP tumors demonstrated higher levels of somatic mutations, inflammatory markers, immune checkpoint effector molecules, and improved relapse-free survival. Differences in drug sensitivity patterns were also observed; notably, KL cells showed increased vulnerability to HSP90-inhibitor therapy. This work provides evidence that co-occurring genomic alterations identify subgroups of KRAS-mutant lung adenocarcinoma with distinct biology and therapeutic vulnerabilities. SIGNIFICANCE Co-occurring genetic alterations in STK11/LKB1, TP53, and CDKN2A/B-the latter coupled with low TTF1 expression-define three major subgroups of KRAS-mutant lung adenocarcinoma with distinct biology, patterns of immune-system engagement, and therapeutic vulnerabilities.


Nature Cell Biology | 2014

An integrin β 3 –KRAS–RalB complex drives tumour stemness and resistance to EGFR inhibition

Laetitia Seguin; Shumei Kato; Aleksandra Franovic; M. Fernanda Camargo; Jacqueline Lesperance; Kathryn C. Elliott; Mayra Yebra; Ainhoa Mielgo; Andrew M. Lowy; Hatim Husain; Tina Cascone; Lixia Diao; Jing Wang; Ignacio I. Wistuba; John V. Heymach; Scott M. Lippman; Jay S. Desgrosellier; Sudarshan Anand; Sara M. Weis; David A. Cheresh

Tumour cells, with stem-like properties, are highly aggressive and often show drug resistance. Here, we reveal that integrin αvβ3 serves as a marker of breast, lung and pancreatic carcinomas with stem-like properties that are highly resistant to receptor tyrosine kinase inhibitors such as erlotinib. This was observed in vitro and in mice bearing patient-derived tumour xenografts or in clinical specimens from lung cancer patients who had progressed on erlotinib. Mechanistically, αvβ3, in the unliganded state, recruits KRAS and RalB to the tumour cell plasma membrane, leading to the activation of TBK1 and NF-κB. In fact, αvβ3 expression and the resulting KRAS–RalB–NF-κB pathway were both necessary and sufficient for tumour initiation, anchorage independence, self-renewal and erlotinib resistance. Pharmacological targeting of this pathway with bortezomib reversed both tumour stemness and erlotinib resistance. These findings not only identify αvβ3 as a marker/driver of carcinoma stemness but also reveal a therapeutic strategy to sensitize such tumours to RTK inhibition.


Nature Biotechnology | 2014

Assessing the clinical utility of cancer genomic and proteomic data across tumor types

Yuan Yuan; Eliezer M. Van Allen; Larsson Omberg; Nikhil Wagle; Ali Amin-Mansour; Artem Sokolov; Lauren Averett Byers; Yanxun Xu; Kenneth R. Hess; Lixia Diao; Leng Han; Xuelin Huang; Michael S. Lawrence; John N. Weinstein; Josh Stuart; Gordon B. Mills; Levi A. Garraway; Adam A. Margolin; Gad Getz; Han Liang

Molecular profiling of tumors promises to advance the clinical management of cancer, but the benefits of integrating molecular data with traditional clinical variables have not been systematically studied. Here we retrospectively predict patient survival using diverse molecular data (somatic copy-number alteration, DNA methylation and mRNA, microRNA and protein expression) from 953 samples of four cancer types from The Cancer Genome Atlas project. We find that incorporating molecular data with clinical variables yields statistically significantly improved predictions (FDR < 0.05) for three cancers but those quantitative gains were limited (2.2–23.9%). Additional analyses revealed little predictive power across tumor types except for one case. In clinically relevant genes, we identified 10,281 somatic alterations across 12 cancer types in 2,928 of 3,277 patients (89.4%), many of which would not be revealed in single-tumor analyses. Our study provides a starting point and resources, including an open-access model evaluation platform, for building reliable prognostic and therapeutic strategies that incorporate molecular data.


Cancer Cell | 2015

The Genomic Landscape and Clinical Relevance of A-to-I RNA Editing in Human Cancers

Leng Han; Lixia Diao; Shuangxing Yu; Xiaoyan Xu; Jie Li; Rui Zhang; Yang Yang; Henrica Maria Johanna Werner; A. Karina Eterovic; Yuan Yuan; Jun Li; Nikitha Nair; Rosalba Minelli; Yiu Huen Tsang; Lydia W.T. Cheung; Kang Jin Jeong; Jason Roszik; Zhenlin Ju; Scott E. Woodman; Yiling Lu; Kenneth L. Scott; Jin Billy Li; Gordon B. Mills; Han Liang

Adenosine-to-inosine (A-to-I) RNA editing is a widespread post-transcriptional mechanism, but its genomic landscape and clinical relevance in cancer have not been investigated systematically. We characterized the global A-to-I RNA editing profiles of 6,236 patient samples of 17 cancer types from The Cancer Genome Atlas and revealed a striking diversity of altered RNA-editing patterns in tumors relative to normal tissues. We identified an appreciable number of clinically relevant editing events, many of which are in noncoding regions. We experimentally demonstrated the effects of several cross-tumor nonsynonymous RNA editing events on cell viability and provide the evidence that RNA editing could selectively affect drug sensitivity. These results highlight RNA editing as an exciting theme for investigating cancer mechanisms, biomarkers, and treatments.


PLOS ONE | 2010

Characteristic Male Urine Microbiomes Associate with Asymptomatic Sexually Transmitted Infection

David E. Nelson; Barbara Van Der Pol; Qunfeng Dong; Kashi Vishwanath Revanna; Baochang Fan; Shraddha Easwaran; Erica Sodergren; George M. Weinstock; Lixia Diao; J. Dennis Fortenberry

Background The microbiome of the male urogenital tract is poorly described but it has been suggested that bacterial colonization of the male urethra might impact risk of sexually transmitted infection (STI). Previous cultivation-dependent studies showed that a variety of non-pathogenic bacteria colonize the urethra but did not thoroughly characterize these microbiomes or establish links between the compositions of urethral microbiomes and STI. Methodology/Findings Here, we used 16S rRNA PCR and sequencing to identify bacteria in urine specimens collected from men who lacked symptoms of urethral inflammation but who differed in status for STI. All of the urine samples contained multiple bacterial genera and many contained taxa that colonize the human vagina. Uncultivated bacteria associated with female genital tract pathology were abundant in specimens from men who had STI. Conclusions Urine microbiomes from men with STI were dominated by fastidious, anaerobic and uncultivated bacteria. The same taxa were rare in STI negative individuals. Our findings suggest that the composition of male urine microbiomes is related to STI.

Collaboration


Dive into the Lixia Diao's collaboration.

Top Co-Authors

Avatar

Jing Wang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lauren Averett Byers

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

John V. Heymach

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ignacio I. Wistuba

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pan Tong

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

John D. Minna

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John N. Weinstein

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Don L. Gibbons

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge