Network


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

Hotspot


Dive into the research topics where Gordon B. Mills is active.

Publication


Featured researches published by Gordon B. Mills.


Genome Research | 2012

Whole-exome sequencing combined with functional genomics reveals novel candidate driver cancer genes in endometrial cancer

Han Liang; Lydia W.T. Cheung; Jie Li; Zhenlin Ju; Shuangxing Yu; Katherine Stemke-Hale; Turgut Dogruluk; Yiling Lu; Xiuping Liu; Chao Gu; Wei Guo; Steven E. Scherer; Hannah Carter; Shannon N. Westin; Mary D. Dyer; Roeland Verhaak; Fan Zhang; Rachel Karchin; Chang Gong Liu; Karen H. Lu; Russell Broaddus; Kenneth L. Scott; Bryan T. Hennessy; Gordon B. Mills

Endometrial cancer is the most common gynecological malignancy, with more than 280,000 cases occurring annually worldwide. Although previous studies have identified important common somatic mutations in endometrial cancer, they have primarily focused on a small set of known cancer genes and have thus provided a limited view of the molecular basis underlying this disease. Here we have developed an integrated systems-biology approach to identifying novel cancer genes contributing to endometrial tumorigenesis. We first performed whole-exome sequencing on 13 endometrial cancers and matched normal samples, systematically identifying somatic alterations with high precision and sensitivity. We then combined bioinformatics prioritization with high-throughput screening (including both shRNA-mediated knockdown and expression of wild-type and mutant constructs) in a highly sensitive cell viability assay. Our results revealed 12 potential driver cancer genes including 10 tumor-suppressor candidates (ARID1A, INHBA, KMO, TTLL5, GRM8, IGFBP3, AKTIP, PHKA2, TRPS1, and WNT11) and two oncogene candidates (ERBB3 and RPS6KC1). The results in the sensor cell line were recapitulated by siRNA-mediated knockdown in endometrial cancer cell lines. Focusing on ARID1A, we integrated mutation profiles with functional proteomics in 222 endometrial cancer samples, demonstrating that ARID1A mutations frequently co-occur with mutations in the phosphatidylinositol 3-kinase (PI3K) pathway and are associated with PI3K pathway activation. siRNA knockdown in endometrial cancer cell lines increased AKT phosphorylation supporting ARID1A as a novel regulator of PI3K pathway activity. Our study presents the first unbiased view of somatic coding mutations in endometrial cancer and provides functional evidence for diverse driver genes and mutations in this disease.


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.


Clinical Cancer Research | 2016

Homologous Recombination Deficiency (HRD) Score Predicts Response to Platinum-Containing Neoadjuvant Chemotherapy in Patients with Triple-Negative Breast Cancer

Melinda L. Telli; Kirsten Timms; Julia Reid; Bryan T. Hennessy; Gordon B. Mills; Kristin C. Jensen; Zoltan Szallasi; William T. Barry; Nadine Tung; Steven J. Isakoff; Paula D. Ryan; April Greene-Colozzi; Alexander Gutin; Zaina Sangale; Diana Iliev; Chris Neff; Victor Abkevich; Joshua Jones; Jerry S. Lanchbury; Anne-Renee Hartman; Judy Garber; James M. Ford; Daniel P. Silver; Andrea L. Richardson

Purpose: BRCA1/2-mutated and some sporadic triple-negative breast cancers (TNBC) have DNA repair defects and are sensitive to DNA-damaging therapeutics. Recently, three independent DNA-based measures of genomic instability were developed on the basis of loss of heterozygosity (LOH), telomeric allelic imbalance (TAI), and large-scale state transitions (LST). Experimental Design: We assessed a combined homologous recombination deficiency (HRD) score, an unweighted sum of LOH, TAI, and LST scores, in three neoadjuvant TNBC trials of platinum-containing therapy. We then tested the association of HR deficiency, defined as HRD score ≥42 or BRCA1/2 mutation, with response to platinum-based therapy. Results: In a trial of neoadjuvant platinum, gemcitabine, and iniparib, HR deficiency predicted residual cancer burden score of 0 or I (RCB 0/I) and pathologic complete response (pCR; OR = 4.96, P = 0.0036; OR = 6.52, P = 0.0058). HR deficiency remained a significant predictor of RCB 0/I when adjusted for clinical variables (OR = 5.86, P = 0.012). In two other trials of neoadjuvant cisplatin therapy, HR deficiency predicted RCB 0/I and pCR (OR = 10.18, P = 0.0011; OR = 17.00, P = 0.0066). In a multivariable model of RCB 0/I, HR deficiency retained significance when clinical variables were included (OR = 12.08, P = 0.0017). When restricted to BRCA1/2 nonmutated tumors, response was higher in patients with high HRD scores: RCB 0/I P = 0.062, pCR P = 0.063 in the neoadjuvant platinum, gemcitabine, and iniparib trial; RCB 0/I P = 0.0039, pCR P = 0.018 in the neoadjuvant cisplatin trials. Conclusions: HR deficiency identifies TNBC tumors, including BRCA1/2 nonmutated tumors more likely to respond to platinum-containing therapy. Clin Cancer Res; 22(15); 3764–73. ©2016 AACR.


Journal of Investigative Dermatology | 2015

Beyond BRAFV600: Clinical Mutation Panel Testing by Next-Generation Sequencing in Advanced Melanoma

Alan E. Siroy; Genevieve M. Boland; Denái R. Milton; Jason Roszik; Silva Frankian; Jared Malke; Lauren E. Haydu; Victor G. Prieto; Michael Tetzlaff; Doina Ivan; Wei Lien Wang; Carlos Torres-Cabala; Jonathan L. Curry; Sinchita Roy-Chowdhuri; Russell R. Broaddus; Asif Rashid; John Stewart; Jeffrey E. Gershenwald; Rodabe N. Amaria; Sapna P. Patel; Nicholas E. Papadopoulos; Agop Bedikian; Wen Jen Hwu; Patrick Hwu; Adi Diab; Scott E. Woodman; Kenneth D. Aldape; Rajyalakshmi Luthra; Keyur Patel; Kenna R. Shaw

The management of melanoma has evolved due to improved understanding of its molecular drivers. To augment the current understanding of the prevalence, patterns, and associations of mutations in this disease, the results of clinical testing of 699 advanced melanoma patients using a pan-cancer next generation sequencing (NGS) panel of hotspot regions in 46 genes were reviewed. Mutations were identified in 43 of the 46 genes on the panel. The most common mutations were BRAFV600 (36%), NRAS (21%), TP53 (16%), BRAFNon-V600 (6%), and KIT (4%). Approximately one-third of melanomas had >1 mutation detected, and the number of mutations per tumor was associated with melanoma subtype. Concurrent TP53 mutations were the most frequent event in tumors with BRAFV600 and NRAS mutations. Melanomas with BRAFNon-V600 mutations frequently harbored concurrent NRAS mutations (18%), which were rare in tumors with BRAFV600 mutations (1.6%). The prevalence of BRAFV600 and KIT mutations were significantly associated with melanoma subtypes, and BRAFV600 and TP53 mutations were significantly associated with cutaneous primary tumor location. Multiple potential therapeutic targets were identified in metastatic unknown primary and cutaneous melanomas that lacked BRAFV600 and NRAS mutations. These results enrich our understanding of the patterns and clinical associations of oncogenic mutations in melanoma.


Science Signaling | 2012

Network analysis of the focal adhesion to invadopodia transition identifies a PI3K-PKCα invasive signaling axis.

Daisuke Hoshino; Jerome Jourquin; Shane Weller Emmons; Tyne Miller; Margalit Goldgof; Kaitlin Costello; Darren R. Tyson; Brandee T. Brown; Yiling Lu; Nagendra K. Prasad; Bing Zhang; Gordon B. Mills; Wendell G. Yarbrough; Vito Quaranta; Motoharu Seiki; Alissa M. Weaver

The activity of the lipid kinase PI3K governs whether the protein kinase PKCα promotes invasive behavior of cancer cells. Becoming Invasive Invasive and metastatic cancer cells form cellular protrusions called invadopodia that can degrade the extracellular matrix. Hoshino et al. integrated data from head and neck carcinomas with network analysis of invadopodia and focal adhesions, cellular structures that contain many of the same components as invadopodia but have decreased ability to degrade the extracellular matrix. They identified phosphatidylinositol 3-kinase (PI3K) and protein kinase C α (PKCα) as key determinants in the formation of invadopodia. The formation of invadopodia was enhanced by PKCα in cells with wild-type PI3K but was inhibited by PKCα in cells with enhanced PI3K activity (due to expression of components of the PI3K pathway with cancer-associated mutations). These results suggested that PKCα participated in a negative feedback loop that limited the activity of PI3K and thus cellular invasiveness, which the authors confirmed. The combination of high PI3K activity with low PKCα activity correlated with increased number of invadopodia in cell lines derived from head and neck carcinoma, breast cancer, or melanoma. This PI3K-high and PKCα-low signaling state may be useful as a biomarker for cancer aggressiveness. In cancer, deregulated signaling can produce an invasive cellular phenotype. We modeled the invasive transition as a theoretical switch between two cytoskeletal structures: focal adhesions and extracellular matrix–degrading invadopodia. We constructed molecular interaction networks of each structure and identified upstream regulatory hubs through computational analyses. We compared these regulatory hubs to the status of signaling components from head and neck carcinomas, which led us to analyze phosphatidylinositol 3-kinase (PI3K) and protein kinase C α (PKCα). Consistent with previous studies, PI3K activity promoted both the formation and the activity of invadopodia. We found that PI3K induction of invadopodia was increased by overexpression of SH2 (Src homology 2) domain–containing inositol 5′-phosphatase 2 (SHIP2), which converts the phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3] that is produced by PI3K activity to phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2], which is believed to promote invadopodia formation. Knockdown of PKCα had divergent effects on invadopodia formation, depending on the status of PI3K. Loss of PKCα inhibited invadopodia formation in cells with wild-type PI3K pathway status. Conversely, in cells with constitutively active PI3K (through activating PI3K mutants or lacking the endogenous opposing enzyme PTEN), PKCα knockdown increased invadopodia formation. Mechanistic studies revealed a negative feedback loop from PKCα that dampened PI3K activity and invasive behavior in cells with genetic hyperactivation of the PI3K pathway. These studies demonstrated the potential of network modeling as a discovery tool and identified PI3K and PKCα as interacting regulators of invasive behavior.


Genome Biology | 2008

A sequence-based survey of the complex structural organization of tumor genomes

Benjamin J. Raphael; Stanislav Volik; Peng Yu; Chunxiao Wu; Guiqing Huang; Elena V. Linardopoulou; Barbara J. Trask; Frederic M. Waldman; Joseph F. Costello; Kenneth J. Pienta; Gordon B. Mills; Krystyna Bajsarowicz; Yasuko Kobayashi; Shivaranjani Sridharan; Pamela L. Paris; Quanzhou Tao; Sarah J. Aerni; Raymond P. Brown; Ali Bashir; Joe W. Gray; Jan Fang Cheng; Pieter J. de Jong; Mikhail Nefedov; Thomas Ried; Hesed Padilla-Nash; Colin Collins

BackgroundThe genomes of many epithelial tumors exhibit extensive chromosomal rearrangements. All classes of genome rearrangements can be identified using end sequencing profiling, which relies on paired-end sequencing of cloned tumor genomes.ResultsIn the present study brain, breast, ovary, and prostate tumors, along with three breast cancer cell lines, were surveyed using end sequencing profiling, yielding the largest available collection of sequence-ready tumor genome breakpoints and providing evidence that some rearrangements may be recurrent. Sequencing and fluorescence in situ hybridization confirmed translocations and complex tumor genome structures that include co-amplification and packaging of disparate genomic loci with associated molecular heterogeneity. Comparison of the tumor genomes suggests recurrent rearrangements. Some are likely to be novel structural polymorphisms, whereas others may be bona fide somatic rearrangements. A recurrent fusion transcript in breast tumors and a constitutional fusion transcript resulting from a segmental duplication were identified. Analysis of end sequences for single nucleotide polymorphisms revealed candidate somatic mutations and an elevated rate of novel single nucleotide polymorphisms in an ovarian tumor.ConclusionThese results suggest that the genomes of many epithelial tumors may be far more dynamic and complex than was previously appreciated and that genomic fusions, including fusion transcripts and proteins, may be common, possibly yielding tumor-specific biomarkers and therapeutic targets.


PLOS ONE | 2011

Selective Genomic Copy Number Imbalances and Probability of Recurrence in Early-Stage Breast Cancer

Patricia A. Thompson; Abenaa Brewster; Do Kim-Anh; Veerabhadran Baladandayuthapani; Bradley M. Broom; Mary E. Edgerton; Karin M. Hahn; James L. Murray; Aysegul Sahin; Spyros Tsavachidis; Yuker Wang; Li Zhang; Gabriel N. Hortobagyi; Gordon B. Mills; Melissa L. Bondy

A number of studies of copy number imbalances (CNIs) in breast tumors support associations between individual CNIs and patient outcomes. However, no pattern or signature of CNIs has emerged for clinical use. We determined copy number (CN) gains and losses using high-density molecular inversion probe (MIP) arrays for 971 stage I/II breast tumors and applied a boosting strategy to fit hazards models for CN and recurrence, treating chromosomal segments in a dose-specific fashion (-1 [loss], 0 [no change] and +1 [gain]). The concordance index (C-Index) was used to compare prognostic accuracy between a training (nu200a=u200a728) and test (nu200a=u200a243) set and across models. Twelve novel prognostic CNIs were identified: losses at 1p12, 12q13.13, 13q12.3, 22q11, and Xp21, and gains at 2p11.1, 3q13.12, 10p11.21, 10q23.1, 11p15, 14q13.2-q13.3, and 17q21.33. In addition, seven CNIs previously implicated as prognostic markers were selected: losses at 8p22 and 16p11.2 and gains at 10p13, 11q13.5, 12p13, 20q13, and Xq28. For all breast cancers combined, the final full model including 19 CNIs, clinical covariates, and tumor marker-approximated subtypes (estrogen receptor [ER], progesterone receptor, ERBB2 amplification, and Ki67) significantly outperformed a model containing only clinical covariates and tumor subtypes (C-Index full model, train[test] u200a=u200a 0.72[0.71] ± 0.02 vs. C-Index clinical + subtype model, train[test] u200a=u200a 0.62[0.62] ± 0.02; p<10−6). In addition, the full model containing 19 CNIs significantly improved prognostication separately for ER–, HER2+, luminal B, and triple negative tumors over clinical variables alone. In summary, we show that a set of 19 CNIs discriminates risk of recurrence among early-stage breast tumors, independent of ER status. Further, our data suggest the presence of specific CNIs that promote and, in some cases, limit tumor spread.


Molecular Cancer Therapeutics | 2016

AZD5153: a novel bivalent BET bromodomain inhibitor highly active against hematologic malignancies

Garrett W. Rhyasen; Maureen Hattersley; Yi Yao; Austin Dulak; Wenxian Wang; Philip Petteruti; Ian L. Dale; Scott Boiko; Tony Cheung; Jingwen Zhang; Shenghua Wen; Lillian Castriotta; Deborah Lawson; Mike Collins; Larry Bao; Miika Ahdesmaki; Graeme Walker; Greg O'Connor; Tammie C. Yeh; Alfred A. Rabow; Jonathan R. Dry; Corinne Reimer; Paul Lyne; Gordon B. Mills; Stephen Fawell; Michael J. Waring; Michael Zinda; Edwin Clark; Huawei Chen

The bromodomain and extraterminal (BET) protein BRD4 regulates gene expression via recruitment of transcriptional regulatory complexes to acetylated chromatin. Pharmacological targeting of BRD4 bromodomains by small molecule inhibitors has proven to be an effective means to disrupt aberrant transcriptional programs critical for tumor growth and/or survival. Herein, we report AZD5153, a potent, selective, and orally available BET/BRD4 bromodomain inhibitor possessing a bivalent binding mode. Unlike previously described monovalent inhibitors, AZD5153 ligates two bromodomains in BRD4 simultaneously. The enhanced avidity afforded through bivalent binding translates into increased cellular and antitumor activity in preclinical hematologic tumor models. In vivo administration of AZD5153 led to tumor stasis or regression in multiple xenograft models of acute myeloid leukemia, multiple myeloma, and diffuse large B-cell lymphoma. The relationship between AZD5153 exposure and efficacy suggests that prolonged BRD4 target coverage is a primary efficacy driver. AZD5153 treatment markedly affects transcriptional programs of MYC, E2F, and mTOR. Of note, mTOR pathway modulation is associated with cell line sensitivity to AZD5153. Transcriptional modulation of MYC and HEXIM1 was confirmed in AZD5153-treated human whole blood, thus supporting their use as clinical pharmacodynamic biomarkers. This study establishes AZD5153 as a highly potent, orally available BET/BRD4 inhibitor and provides a rationale for clinical development in hematologic malignancies. Mol Cancer Ther; 15(11); 2563–74. ©2016 AACR.


Cancer Cell | 2017

Characterization of Human Cancer Cell Lines by Reverse-phase Protein Arrays

Jun Li; Wei Zhao; Rehan Akbani; Wenbin Liu; Zhenlin Ju; Shiyun Ling; Christopher P. Vellano; Paul Roebuck; Qinghua Yu; A. Karina Eterovic; Lauren Averett Byers; Michael A. Davies; Wanleng Deng; Y.N. Vashisht Gopal; Guo Chen; Erika von Euw; Dennis J. Slamon; Dylan Conklin; John V. Heymach; Adi F. Gazdar; John D. Minna; Jeffrey N. Myers; Yiling Lu; Gordon B. Mills; Han Liang

Cancer cell lines are major model systems for mechanistic investigation and drug development. However, protein expression data linked to high-quality DNA, RNA, and drug-screening data have not been available across a large number of cancer cell lines. Using reverse-phase protein arrays, we measured expression levels of ∼230 key cancer-related proteins in >650 independent cell lines, many of which have publically available genomic, transcriptomic, and drug-screening data. Our dataset recapitulates the effects of mutated pathways on protein expression observed in patient samples, and demonstrates that proteins and particularly phosphoproteins provide information for predicting drug sensitivity that is not available from the corresponding mRNAs. We also developed a user-friendly bioinformatic resource, MCLP, to help serve the biomedical research community.


Molecular Cancer Therapeutics | 2016

BRAF Mutation Testing in Cell-Free DNA from the Plasma of Patients with Advanced Cancers Using a Rapid, Automated Molecular Diagnostics System

Filip Janku; Helen J. Huang; Bart Claes; Gerald S. Falchook; Siqing Fu; David S. Hong; Nishma M. Ramzanali; Giovanni Nitti; Goran Cabrilo; Apostolia M. Tsimberidou; Aung Naing; Sarina Anne Piha-Paul; Jennifer J. Wheler; Daniel D. Karp; Veronica R. Holley; Ralph Zinner; Vivek Subbiah; Rajyalakshmi Luthra; Scott Kopetz; Michael J. Overman; Bryan K. Kee; Sapna Pradyuman Patel; Benoit Devogelaere; Erwin Sablon; Geert Maertens; Gordon B. Mills; Razelle Kurzrock; Funda Meric-Bernstam

Cell-free (cf) DNA from plasma offers an easily obtainable material for BRAF mutation analysis for diagnostics and response monitoring. In this study, plasma-derived cfDNA samples from patients with progressing advanced cancers or malignant histiocytosis with known BRAFV600 status from formalin-fixed paraffin-embedded (FFPE) tumors were tested using a prototype version of the Idylla BRAF Mutation Test, a fully integrated real-time PCR-based test with turnaround time about 90 minutes. Of 160 patients, BRAFV600 mutations were detected in 62 (39%) archival FFPE tumor samples and 47 (29%) plasma cfDNA samples. The two methods had overall agreement in 141 patients [88%; κ, 0.74; SE, 0.06; 95% confidence interval (CI), 0.63–0.85]. Idylla had a sensitivity of 73% (95% CI, 0.60–0.83) and specificity of 98% (95% CI, 0.93–1.00). A higher percentage, but not concentration, of BRAFV600 cfDNA in the wild-type background (>2% vs. ≤ 2%) was associated with shorter overall survival (OS; P = 0.005) and in patients with BRAF mutations in the tissue, who were receiving BRAF/MEK inhibitors, shorter time to treatment failure (TTF; P = 0.001). Longitudinal monitoring demonstrated that decreasing levels of BRAFV600 cfDNA were associated with longer TTF (P = 0.045). In conclusion, testing for BRAFV600 mutations in plasma cfDNA using the Idylla BRAF Mutation Test has acceptable concordance with standard testing of tumor tissue. A higher percentage of mutant BRAFV600 in cfDNA corresponded with shorter OS and in patients receiving BRAF/MEK inhibitors also with shorter TTF. Mol Cancer Ther; 15(6); 1397–404. ©2016 AACR.

Collaboration


Dive into the Gordon B. Mills's collaboration.

Top Co-Authors

Avatar

Yiling Lu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Zhenlin Ju

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher P. Vellano

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Funda Meric-Bernstam

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Han Liang

University of Texas System

View shared research outputs
Top Co-Authors

Avatar

Kang Jin Jeong

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Aysegul A. Sahin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

John Mendelsohn

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jun Li

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kenneth L. Scott

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge