501
|
Geeleher P, Cox NJ, Huang RS. Cancer biomarker discovery is improved by accounting for variability in general levels of drug sensitivity in pre-clinical models. Genome Biol 2016; 17:190. [PMID: 27654937 PMCID: PMC5031330 DOI: 10.1186/s13059-016-1050-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/31/2016] [Indexed: 02/02/2023] Open
Abstract
We show that variability in general levels of drug sensitivity in pre-clinical cancer models confounds biomarker discovery. However, using a very large panel of cell lines, each treated with many drugs, we could estimate a general level of sensitivity to all drugs in each cell line. By conditioning on this variable, biomarkers were identified that were more likely to be effective in clinical trials than those identified using a conventional uncorrected approach. We find that differences in general levels of drug sensitivity are driven by biologically relevant processes. We developed a gene expression based method that can be used to correct for this confounder in future studies.
Collapse
Affiliation(s)
- Paul Geeleher
- Section of Hematology/Oncology, The University of Chicago, 900 E 57th Street, KCBD room 7148, Chicago, IL, 60637, USA
| | - Nancy J Cox
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA.,Division of Genetic Medicine, Vanderbilt University, Nashville, TN, USA
| | - R Stephanie Huang
- Section of Hematology/Oncology, The University of Chicago, 900 E 57th Street, KCBD room 7148, Chicago, IL, 60637, USA.
| |
Collapse
|
502
|
Uitdehaag JCM, de Roos JADM, Prinsen MBW, Willemsen-Seegers N, de Vetter JRF, Dylus J, van Doornmalen AM, Kooijman J, Sawa M, van Gerwen SJC, de Man J, Buijsman RC, Zaman GJR. Cell Panel Profiling Reveals Conserved Therapeutic Clusters and Differentiates the Mechanism of Action of Different PI3K/mTOR, Aurora Kinase and EZH2 Inhibitors. Mol Cancer Ther 2016; 15:3097-3109. [PMID: 27587489 DOI: 10.1158/1535-7163.mct-16-0403] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/08/2016] [Accepted: 08/21/2016] [Indexed: 11/16/2022]
Abstract
Cancer cell line panels are important tools to characterize the in vitro activity of new investigational drugs. Here, we present the inhibition profiles of 122 anticancer agents in proliferation assays with 44 or 66 genetically characterized cancer cell lines from diverse tumor tissues (Oncolines). The library includes 29 cytotoxics, 68 kinase inhibitors, and 11 epigenetic modulators. For 38 compounds this is the first comparative profiling in a cell line panel. By strictly maintaining optimized assay protocols, biological variation was kept to a minimum. Replicate profiles of 16 agents over three years show a high average Pearson correlation of 0.8 using IC50 values and 0.9 using GI50 values. Good correlations were observed with other panels. Curve fitting appears a large source of variation. Hierarchical clustering revealed 44 basic clusters, of which 26 contain compounds with common mechanisms of action, of which 9 were not reported before, including TTK, BET and two clusters of EZH2 inhibitors. To investigate unexpected clusterings, sets of BTK, Aurora and PI3K inhibitors were profiled in biochemical enzyme activity assays and surface plasmon resonance binding assays. The BTK inhibitor ibrutinib clusters with EGFR inhibitors, because it cross-reacts with EGFR. Aurora kinase inhibitors separate into two clusters, related to Aurora A or pan-Aurora selectivity. Similarly, 12 inhibitors in the PI3K/AKT/mTOR pathway separated into different clusters, reflecting biochemical selectivity (pan-PI3K, PI3Kβγδ-isoform selective or mTOR-selective). Of these, only allosteric mTOR inhibitors preferentially targeted PTEN-mutated cell lines. This shows that cell line profiling is an excellent tool for the unbiased classification of antiproliferative compounds. Mol Cancer Ther; 15(12); 3097-109. ©2016 AACR.
Collapse
Affiliation(s)
- Joost C M Uitdehaag
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | | | - Martine B W Prinsen
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | | | | | - Jelle Dylus
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | | | - Jeffrey Kooijman
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | | | | | - Jos de Man
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | - Rogier C Buijsman
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | - Guido J R Zaman
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands.
| |
Collapse
|
503
|
Zänker KS, Borresen-Dale AL, Huber HP. Personalized Cancer Care: Risk Prediction, Early Diagnosis, Progression, and Therapy. Biomed Hub 2016; 1:1-9. [PMID: 31988890 PMCID: PMC6945940 DOI: 10.1159/000453253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 10/25/2016] [Indexed: 12/12/2022] Open
Abstract
At the annual prestigious International Symposium of the Fritz-Bender Foundation, Munich, 18-20 May, 2016, researchers, clinicians, and students discussed the state of the art and future perspectives of genomic medicine in cancer. Genomic medicine (also known as precision medicine/oncology) should help clinicians to provide a more precise diagnosis and therapy in oncology for individual patients. The meeting focused on next-generation sequencing methods, analytical computational analysis of big data, and data mining on the way to translational and evidence-based medicine. The meeting covered the social and ethical impact of genomic medicine as well as news and views on antibody targeting of intracellular proteins, on the architecture of intracellular proteins and their impact on carcinogenesis, and on the adaptation of tumor therapy in due consideration of tumor evolution. Subheadings like "Genetic Profiling of Patients and Risk Prediction," "Molecular Profiling of Tumors and Metastases," "Tumor-Host Microenvironment Interaction and Metabolism," and "Targeted Therapy" were subsumed under the main heading of "Personalized Cancer Care."
Collapse
Affiliation(s)
- Kurt S. Zänker
- Institute of Immunology and Experimental Oncology, Center for Biomedical Education and Research, University Witten/Herdecke, Witten, Germany
| | - Anne-Lise Borresen-Dale
- Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
504
|
Serra-Musach J, Mateo F, Capdevila-Busquets E, de Garibay GR, Zhang X, Guha R, Thomas CJ, Grueso J, Villanueva A, Jaeger S, Heyn H, Vizoso M, Pérez H, Cordero A, Gonzalez-Suarez E, Esteller M, Moreno-Bueno G, Tjärnberg A, Lázaro C, Serra V, Arribas J, Benson M, Gustafsson M, Ferrer M, Aloy P, Pujana MÀ. Cancer network activity associated with therapeutic response and synergism. Genome Med 2016; 8:88. [PMID: 27553366 PMCID: PMC4995628 DOI: 10.1186/s13073-016-0340-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 08/01/2016] [Indexed: 12/14/2022] Open
Abstract
Background Cancer patients often show no or only modest benefit from a given therapy. This major problem in oncology is generally attributed to the lack of specific predictive biomarkers, yet a global measure of cancer cell activity may support a comprehensive mechanistic understanding of therapy efficacy. We reasoned that network analysis of omic data could help to achieve this goal. Methods A measure of “cancer network activity” (CNA) was implemented based on a previously defined network feature of communicability. The network nodes and edges corresponded to human proteins and experimentally identified interactions, respectively. The edges were weighted proportionally to the expression of the genes encoding for the corresponding proteins and relative to the number of direct interactors. The gene expression data corresponded to the basal conditions of 595 human cancer cell lines. Therapeutic responses corresponded to the impairment of cell viability measured by the half maximal inhibitory concentration (IC50) of 130 drugs approved or under clinical development. Gene ontology, signaling pathway, and transcription factor-binding annotations were taken from public repositories. Predicted synergies were assessed by determining the viability of four breast cancer cell lines and by applying two different analytical methods. Results The effects of drug classes were associated with CNAs formed by different cell lines. CNAs also differentiate target families and effector pathways. Proteins that occupy a central position in the network largely contribute to CNA. Known key cancer-associated biological processes, signaling pathways, and master regulators also contribute to CNA. Moreover, the major cancer drivers frequently mediate CNA and therapeutic differences. Cell-based assays centered on these differences and using uncorrelated drug effects reveals novel synergistic combinations for the treatment of breast cancer dependent on PI3K-mTOR signaling. Conclusions Cancer therapeutic responses can be predicted on the basis of a systems-level analysis of molecular interactions and gene expression. Fundamental cancer processes, pathways, and drivers contribute to this feature, which can also be exploited to predict precise synergistic drug combinations. Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0340-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jordi Serra-Musach
- Breast Cancer and Systems Biology Lab, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Francesca Mateo
- Breast Cancer and Systems Biology Lab, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Eva Capdevila-Busquets
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, Barcelona, 08028, Catalonia, Spain
| | - Gorka Ruiz de Garibay
- Breast Cancer and Systems Biology Lab, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Dr. Rockville, Bethesda, MD, 20850, USA
| | - Raj Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Dr. Rockville, Bethesda, MD, 20850, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Dr. Rockville, Bethesda, MD, 20850, USA
| | - Judit Grueso
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology (VHIO), Cellex Center, Natzaret 115-117, Barcelona, 08035, Catalonia, Spain
| | - Alberto Villanueva
- Breast Cancer and Systems Biology Lab, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Samira Jaeger
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, Barcelona, 08028, Catalonia, Spain
| | - Holger Heyn
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Miguel Vizoso
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Hector Pérez
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Alex Cordero
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Eva Gonzalez-Suarez
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain.,Department of Physiological Sciences II, School of Medicine, University of Barcelona, Feixa Llarga s/n, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, Barcelona, 08010, Catalonia, Spain
| | - Gema Moreno-Bueno
- Department of Biochemistry, Autonomous University of Madrid (UAM), Biomedical Research Institute "Alberto Sols" (Spanish National Research Council (CSIC)-UAM), Hospital La Paz Institute for Health Research (IdiPAZ), Arzobispo Morcillo 4, Madrid, 28029, Spain.,MD Anderson International Foundation, Arturo Soria 270, Madrid, 28033, Spain
| | - Andreas Tjärnberg
- The Centre for Individualized Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, 58183, Sweden
| | - Conxi Lázaro
- Hereditary Cancer Program, ICO, IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology (VHIO), Cellex Center, Natzaret 115-117, Barcelona, 08035, Catalonia, Spain
| | - Joaquín Arribas
- Catalan Institution for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, Barcelona, 08010, Catalonia, Spain.,Preclinical Research Program, VHIO, Cellex Center, Natzaret 115-117, Barcelona, 08035, Catalonia, Spain.,Department of Biochemistry and Molecular Biology, Medical School Building M, Autonomous University of Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - Mikael Benson
- The Centre for Individualized Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, 58183, Sweden
| | - Mika Gustafsson
- The Centre for Individualized Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, 58183, Sweden
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Dr. Rockville, Bethesda, MD, 20850, USA.
| | - Patrick Aloy
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, Barcelona, 08028, Catalonia, Spain. .,Catalan Institution for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, Barcelona, 08010, Catalonia, Spain.
| | - Miquel Àngel Pujana
- Breast Cancer and Systems Biology Lab, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain.
| |
Collapse
|
505
|
Abstract
The major challenge underlying the emerging precision medicine initiative is to make links between cancer subsets and drugs that can be used to guide treatment of individual patients, leading to improved outcomes and decreased toxicity. Seashore-Ludlow and colleagues support this effort by reporting measurements of responses of 664 adherent cancer cell lines to 70 FDA-approved drugs, 100 experimental compounds, and 311 small-molecule probes. They use a novel Annotated Cluster Multidimensional Enrichment algorithm to identify drug mechanisms of action, molecular markers of response, responsive cancer subtypes, and compounds that produce synergistic cell inhibition.
Collapse
Affiliation(s)
- Joe W Gray
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon.
| | - Gordon B Mills
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
506
|
Pozdeyev N, Yoo M, Mackie R, Schweppe RE, Tan AC, Haugen BR. Integrating heterogeneous drug sensitivity data from cancer pharmacogenomic studies. Oncotarget 2016; 7:51619-51625. [PMID: 27322211 PMCID: PMC5239501 DOI: 10.18632/oncotarget.10010] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/29/2016] [Indexed: 01/22/2023] Open
Abstract
The consistency of in vitro drug sensitivity data is of key importance for cancer pharmacogenomics. Previous attempts to correlate drug sensitivities from the large pharmacogenomics databases, such as the Cancer Cell Line Encyclopedia (CCLE) and the Genomics of Drug Sensitivity in Cancer (GDSC), have produced discordant results. We developed a new drug sensitivity metric, the area under the dose response curve adjusted for the range of tested drug concentrations, which allows integration of heterogeneous drug sensitivity data from the CCLE, the GDSC, and the Cancer Therapeutics Response Portal (CTRP). We show that there is moderate to good agreement of drug sensitivity data for many targeted therapies, particularly kinase inhibitors. The results of this largest cancer cell line drug sensitivity data analysis to date are accessible through the online portal, which serves as a platform for high power pharmacogenomics analysis.
Collapse
Affiliation(s)
- Nikita Pozdeyev
- Department of Medicine, University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Minjae Yoo
- Department of Medicine, University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ryan Mackie
- Department of Medicine, University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Rebecca E. Schweppe
- Department of Medicine, University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Aik Choon Tan
- Department of Medicine, University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Bryan R. Haugen
- Department of Medicine, University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
507
|
Optimized design and analysis of preclinical intervention studies in vivo. Sci Rep 2016; 6:30723. [PMID: 27480578 PMCID: PMC4969752 DOI: 10.1038/srep30723] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 07/06/2016] [Indexed: 12/20/2022] Open
Abstract
Recent reports have called into question the reproducibility, validity and translatability of the preclinical animal studies due to limitations in their experimental design and statistical analysis. To this end, we implemented a matching-based modelling approach for optimal intervention group allocation, randomization and power calculations, which takes full account of the complex animal characteristics at baseline prior to interventions. In prostate cancer xenograft studies, the method effectively normalized the confounding baseline variability, and resulted in animal allocations which were supported by RNA-seq profiling of the individual tumours. The matching information increased the statistical power to detect true treatment effects at smaller sample sizes in two castration-resistant prostate cancer models, thereby leading to saving of both animal lives and research costs. The novel modelling approach and its open-source and web-based software implementations enable the researchers to conduct adequately-powered and fully-blinded preclinical intervention studies, with the aim to accelerate the discovery of new therapeutic interventions.
Collapse
|
508
|
Hanaford AR, Archer TC, Price A, Kahlert UD, Maciaczyk J, Nikkhah G, Kim JW, Ehrenberger T, Clemons PA, Dančík V, Seashore-Ludlow B, Viswanathan V, Stewart ML, Rees MG, Shamji A, Schreiber S, Fraenkel E, Pomeroy SL, Mesirov JP, Tamayo P, Eberhart CG, Raabe EH. DiSCoVERing Innovative Therapies for Rare Tumors: Combining Genetically Accurate Disease Models with In Silico Analysis to Identify Novel Therapeutic Targets. Clin Cancer Res 2016; 22:3903-14. [PMID: 27012813 PMCID: PMC5055054 DOI: 10.1158/1078-0432.ccr-15-3011] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/10/2016] [Indexed: 12/20/2022]
Abstract
PURPOSE We used human stem and progenitor cells to develop a genetically accurate novel model of MYC-driven Group 3 medulloblastoma. We also developed a new informatics method, Disease-model Signature versus Compound-Variety Enriched Response ("DiSCoVER"), to identify novel therapeutics that target this specific disease subtype. EXPERIMENTAL DESIGN Human neural stem and progenitor cells derived from the cerebellar anlage were transduced with oncogenic elements associated with aggressive medulloblastoma. An in silico analysis method for screening drug sensitivity databases (DiSCoVER) was used in multiple drug sensitivity datasets. We validated the top hits from this analysis in vitro and in vivo RESULTS Human neural stem and progenitor cells transformed with c-MYC, dominant-negative p53, constitutively active AKT and hTERT formed tumors in mice that recapitulated Group 3 medulloblastoma in terms of pathology and expression profile. DiSCoVER analysis predicted that aggressive MYC-driven Group 3 medulloblastoma would be sensitive to cyclin-dependent kinase (CDK) inhibitors. The CDK 4/6 inhibitor palbociclib decreased proliferation, increased apoptosis, and significantly extended the survival of mice with orthotopic medulloblastoma xenografts. CONCLUSIONS We present a new method to generate genetically accurate models of rare tumors, and a companion computational methodology to find therapeutic interventions that target them. We validated our human neural stem cell model of MYC-driven Group 3 medulloblastoma and showed that CDK 4/6 inhibitors are active against this subgroup. Our results suggest that palbociclib is a potential effective treatment for poor prognosis MYC-driven Group 3 medulloblastoma tumors in carefully selected patients. Clin Cancer Res; 22(15); 3903-14. ©2016 AACR.
Collapse
Affiliation(s)
- Allison R Hanaford
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Tenley C Archer
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, Massachusetts. Department of Neurology, Boston Children's Hospital, Harvard Medical School, Cambridge, Massachusetts
| | - Antoinette Price
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ulf D Kahlert
- Department of Neurosurgery, Heinrich-Heine University Hospital, Duesseldorf, Germany
| | - Jarek Maciaczyk
- Department of Neurosurgery, Heinrich-Heine University Hospital, Duesseldorf, Germany
| | - Guido Nikkhah
- Department of Neurosurgery, University Hospital, Stuttgart, Germany
| | - Jong Wook Kim
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, Massachusetts. Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tobias Ehrenberger
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, Massachusetts. Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Paul A Clemons
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, Massachusetts. Center for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts
| | - Vlado Dančík
- Center for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts
| | | | - Vasanthi Viswanathan
- Center for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts
| | - Michelle L Stewart
- Center for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts
| | - Matthew G Rees
- Center for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts
| | - Alykhan Shamji
- Center for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts
| | - Stuart Schreiber
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, Massachusetts. Center for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts. Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts. Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Ernest Fraenkel
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, Massachusetts. Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Scott L Pomeroy
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, Massachusetts. Department of Neurology, Boston Children's Hospital, Harvard Medical School, Cambridge, Massachusetts
| | - Jill P Mesirov
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, Massachusetts. Department of Medicine, University of California San Diego, La Jolla, California. Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Pablo Tamayo
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, Massachusetts. Department of Medicine, University of California San Diego, La Jolla, California. Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland.
| | - Eric H Raabe
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland. Division of Pediatric Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
509
|
Iorio F, Knijnenburg TA, Vis DJ, Bignell GR, Menden MP, Schubert M, Aben N, Gonçalves E, Barthorpe S, Lightfoot H, Cokelaer T, Greninger P, van Dyk E, Chang H, de Silva H, Heyn H, Deng X, Egan RK, Liu Q, Mironenko T, Mitropoulos X, Richardson L, Wang J, Zhang T, Moran S, Sayols S, Soleimani M, Tamborero D, Lopez-Bigas N, Ross-Macdonald P, Esteller M, Gray NS, Haber DA, Stratton MR, Benes CH, Wessels LFA, Saez-Rodriguez J, McDermott U, Garnett MJ. A Landscape of Pharmacogenomic Interactions in Cancer. Cell 2016; 166:740-754. [PMID: 27397505 PMCID: PMC4967469 DOI: 10.1016/j.cell.2016.06.017] [Citation(s) in RCA: 1284] [Impact Index Per Article: 142.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/23/2015] [Accepted: 06/03/2016] [Indexed: 12/31/2022]
Abstract
Systematic studies of cancer genomes have provided unprecedented insights into the molecular nature of cancer. Using this information to guide the development and application of therapies in the clinic is challenging. Here, we report how cancer-driven alterations identified in 11,289 tumors from 29 tissues (integrating somatic mutations, copy number alterations, DNA methylation, and gene expression) can be mapped onto 1,001 molecularly annotated human cancer cell lines and correlated with sensitivity to 265 drugs. We find that cell lines faithfully recapitulate oncogenic alterations identified in tumors, find that many of these associate with drug sensitivity/resistance, and highlight the importance of tissue lineage in mediating drug response. Logic-based modeling uncovers combinations of alterations that sensitize to drugs, while machine learning demonstrates the relative importance of different data types in predicting drug response. Our analysis and datasets are rich resources to link genotypes with cellular phenotypes and to identify therapeutic options for selected cancer sub-populations. We integrate heterogeneous molecular data of 11,289 tumors and 1,001 cell lines We measure the response of 1,001 cancer cell lines to 265 anti-cancer drugs We uncover numerous oncogenic aberrations that sensitize to an anti-cancer drug Our study forms a resource to identify therapeutic options for cancer sub-populations
Collapse
Affiliation(s)
- Francesco Iorio
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK; Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Theo A Knijnenburg
- Institute for Systems Biology, Seattle, WA 98109, USA; Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Daniel J Vis
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Graham R Bignell
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Michael P Menden
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK; Faculty of Medicine, Joint Research Centre for Computational Biomedicine, RWTH Aachen University, Aachen 52057, Germany
| | - Michael Schubert
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Nanne Aben
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands; Department of EEMCS, Delft University of Technology, Delft 2628 CD, the Netherlands
| | - Emanuel Gonçalves
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Syd Barthorpe
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Howard Lightfoot
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Thomas Cokelaer
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK; Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Patricia Greninger
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Ewald van Dyk
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Han Chang
- Genetically Defined Diseases and Genomics, Bristol-Myers Squibb Research and Development, Hopewell, NJ 08534, USA
| | - Heshani de Silva
- Genetically Defined Diseases and Genomics, Bristol-Myers Squibb Research and Development, Hopewell, NJ 08534, USA
| | - Holger Heyn
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet 08908, Barcelona, Catalonia, Spain
| | - Xianming Deng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Regina K Egan
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Qingsong Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Tatiana Mironenko
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Xeni Mitropoulos
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Laura Richardson
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Tinghu Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Sebastian Moran
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet 08908, Barcelona, Catalonia, Spain
| | - Sergi Sayols
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet 08908, Barcelona, Catalonia, Spain
| | - Maryam Soleimani
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - David Tamborero
- Research Program on Biomedical Informatics, IMIM Hospital del Mar Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Nuria Lopez-Bigas
- Research Program on Biomedical Informatics, IMIM Hospital del Mar Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Catalonia, Spain
| | - Petra Ross-Macdonald
- Genetically Defined Diseases and Genomics, Bristol-Myers Squibb Research and Development, Hopewell, NJ 08534, USA
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet 08908, Barcelona, Catalonia, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Catalonia, Spain; Department of Physiological Sciences II of the School of Medicine, University of Barcelona, L'Hospitalet 08908, Barcelona, Catalonia, Spain
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Daniel A Haber
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Michael R Stratton
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Cyril H Benes
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands; Department of EEMCS, Delft University of Technology, Delft 2628 CD, the Netherlands; Cancer Genomics Netherlands, Uppsalalaan 8, Utrecht 3584CT, the Netherlands
| | - Julio Saez-Rodriguez
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK; Faculty of Medicine, Joint Research Centre for Computational Biomedicine, RWTH Aachen University, Aachen 52057, Germany
| | - Ultan McDermott
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK.
| | - Mathew J Garnett
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK.
| |
Collapse
|
510
|
|
511
|
Hong AL, Tseng YY, Cowley GS, Jonas O, Cheah JH, Kynnap BD, Doshi MB, Oh C, Meyer SC, Church AJ, Gill S, Bielski CM, Keskula P, Imamovic A, Howell S, Kryukov GV, Clemons PA, Tsherniak A, Vazquez F, Crompton BD, Shamji AF, Rodriguez-Galindo C, Janeway KA, Roberts CWM, Stegmaier K, van Hummelen P, Cima MJ, Langer RS, Garraway LA, Schreiber SL, Root DE, Hahn WC, Boehm JS. Integrated genetic and pharmacologic interrogation of rare cancers. Nat Commun 2016; 7:11987. [PMID: 27329820 PMCID: PMC4917959 DOI: 10.1038/ncomms11987] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/18/2016] [Indexed: 02/06/2023] Open
Abstract
Identifying therapeutic targets in rare cancers remains challenging due to the paucity of established models to perform preclinical studies. As a proof-of-concept, we developed a patient-derived cancer cell line, CLF-PED-015-T, from a paediatric patient with a rare undifferentiated sarcoma. Here, we confirm that this cell line recapitulates the histology and harbours the majority of the somatic genetic alterations found in a metastatic lesion isolated at first relapse. We then perform pooled CRISPR-Cas9 and RNAi loss-of-function screens and a small-molecule screen focused on druggable cancer targets. Integrating these three complementary and orthogonal methods, we identify CDK4 and XPO1 as potential therapeutic targets in this cancer, which has no known alterations in these genes. These observations establish an approach that integrates new patient-derived models, functional genomics and chemical screens to facilitate the discovery of targets in rare cancers.
Collapse
Affiliation(s)
- Andrew L. Hong
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Yuen-Yi Tseng
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Glenn S. Cowley
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Oliver Jonas
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Jaime H. Cheah
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Bryan D. Kynnap
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Mihir B. Doshi
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Coyin Oh
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Stephanie C. Meyer
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Alanna J. Church
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - Shubhroz Gill
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Craig M. Bielski
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Paula Keskula
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Alma Imamovic
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Sara Howell
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Gregory V. Kryukov
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA
| | - Paul A. Clemons
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Aviad Tsherniak
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Francisca Vazquez
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Brian D. Crompton
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Alykhan F. Shamji
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Carlos Rodriguez-Galindo
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Katherine A. Janeway
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Charles W. M. Roberts
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Kimberly Stegmaier
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Paul van Hummelen
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Michael J. Cima
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Robert S. Langer
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Levi A. Garraway
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - Stuart L. Schreiber
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - David E. Root
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - William C. Hahn
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA
| | - Jesse S. Boehm
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
512
|
Affiliation(s)
- Adam C Palmer
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, Massachusetts, USA, and the School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
513
|
Hafner M, Niepel M, Chung M, Sorger PK. Growth rate inhibition metrics correct for confounders in measuring sensitivity to cancer drugs. Nat Methods 2016; 13:521-7. [PMID: 27135972 PMCID: PMC4887336 DOI: 10.1038/nmeth.3853] [Citation(s) in RCA: 419] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/01/2016] [Indexed: 12/18/2022]
Abstract
Drug sensitivity and resistance are conventionally quantified by IC50 or Emax values, but these metrics are highly sensitive to the number of divisions taking place over the course of a response assay. The dependency of IC50 and Emax on division rate creates artefactual correlations between genotype and drug sensitivity, while obscuring valuable biological insights and interfering with biomarker discovery. We derive alternative small molecule drug-response metrics that are insensitive to division number. These are based on estimation of the magnitude of drug-induced growth rate inhibition (GR) using endpoint or time-course assays. We show that GR50 and GRmax are superior to conventional metrics for assessing the effects of small molecule drugs in dividing cells. Moreover, adopting GR metrics requires only modest changes in experimental protocols. We expect GR metrics to improve the study of cell signaling and growth using small molecules and biologics and to facilitate the discovery of drug-response biomarkers and the identification of drugs effective against specific patient-derived tumor cells.
Collapse
Affiliation(s)
- Marc Hafner
- HMS LINCS Center, Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Mario Niepel
- HMS LINCS Center, Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Mirra Chung
- HMS LINCS Center, Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Peter K. Sorger
- HMS LINCS Center, Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
514
|
An unbiased metric of antiproliferative drug effect in vitro. Nat Methods 2016; 13:497-500. [PMID: 27135974 PMCID: PMC4887341 DOI: 10.1038/nmeth.3852] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 04/01/2016] [Indexed: 01/15/2023]
Abstract
In vitro cell proliferation assays are widely used in pharmacology, molecular biology, and drug discovery. Using theoretical modeling and experimentation, we show that current metrics of antiproliferative small molecule effect suffer from time-dependent bias, leading to inaccurate assessments of parameters such as drug potency and efficacy. We propose the drug-induced proliferation (DIP) rate, the slope of the line on a plot of cell population doublings versus time, as an alternative, time-independent metric.
Collapse
|
515
|
Yard BD, Adams DJ, Chie EK, Tamayo P, Battaglia JS, Gopal P, Rogacki K, Pearson BE, Phillips J, Raymond DP, Pennell NA, Almeida F, Cheah JH, Clemons PA, Shamji A, Peacock CD, Schreiber SL, Hammerman PS, Abazeed ME. A genetic basis for the variation in the vulnerability of cancer to DNA damage. Nat Commun 2016; 7:11428. [PMID: 27109210 PMCID: PMC4848553 DOI: 10.1038/ncomms11428] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 03/24/2016] [Indexed: 12/22/2022] Open
Abstract
Radiotherapy is not currently informed by the genetic composition of an individual patient's tumour. To identify genetic features regulating survival after DNA damage, here we conduct large-scale profiling of cellular survival after exposure to radiation in a diverse collection of 533 genetically annotated human tumour cell lines. We show that sensitivity to radiation is characterized by significant variation across and within lineages. We combine results from our platform with genomic features to identify parameters that predict radiation sensitivity. We identify somatic copy number alterations, gene mutations and the basal expression of individual genes and gene sets that correlate with the radiation survival, revealing new insights into the genetic basis of tumour cellular response to DNA damage. These results demonstrate the diversity of tumour cellular response to ionizing radiation and establish multiple lines of evidence that new genetic features regulating cellular response after DNA damage can be identified.
Collapse
Affiliation(s)
- Brian D Yard
- Department of Translational Hematology Oncology Research, Cleveland Clinic, 9500 Euclid Avenue/R40, Cleveland, Ohio 44195, USA
| | - Drew J Adams
- Department of Genetics, Case Western Reserve University, 2109 Adelbert Road/BRB, Cleveland, Ohio 44106, USA
| | - Eui Kyu Chie
- Department of Translational Hematology Oncology Research, Cleveland Clinic, 9500 Euclid Avenue/R40, Cleveland, Ohio 44195, USA.,Department of Radiation Oncology, Seoul National University College of Medicine, 101, Daehak-Ro, Jongno-Gu, Seoul 110-774, Korea
| | - Pablo Tamayo
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Jessica S Battaglia
- Department of Translational Hematology Oncology Research, Cleveland Clinic, 9500 Euclid Avenue/R40, Cleveland, Ohio 44195, USA
| | - Priyanka Gopal
- Department of Translational Hematology Oncology Research, Cleveland Clinic, 9500 Euclid Avenue/R40, Cleveland, Ohio 44195, USA
| | - Kevin Rogacki
- Department of Translational Hematology Oncology Research, Cleveland Clinic, 9500 Euclid Avenue/R40, Cleveland, Ohio 44195, USA
| | - Bradley E Pearson
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - James Phillips
- Department of Translational Hematology Oncology Research, Cleveland Clinic, 9500 Euclid Avenue/R40, Cleveland, Ohio 44195, USA
| | - Daniel P Raymond
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, 9500 Euclid Avenue/J4-1, Cleveland, Ohio 44195, USA
| | - Nathan A Pennell
- Department of Hematology and Medical Oncology, Cleveland Clinic, 9500 Euclid Avenue/R40, Cleveland, Ohio 44195, USA
| | - Francisco Almeida
- Department of Pulmonary Medicine, Cleveland Clinic, 9500 Euclid Avenue/M2-141, Cleveland, Ohio 44195, USA
| | - Jaime H Cheah
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, USA.,Center for the Science of Therapeutics, Broad Institute, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Paul A Clemons
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, USA.,Center for the Science of Therapeutics, Broad Institute, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Alykhan Shamji
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, USA.,Center for the Science of Therapeutics, Broad Institute, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Craig D Peacock
- Department of Translational Hematology Oncology Research, Cleveland Clinic, 9500 Euclid Avenue/R40, Cleveland, Ohio 44195, USA
| | - Stuart L Schreiber
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, USA.,Center for the Science of Therapeutics, Broad Institute, 415 Main Street, Cambridge, Massachusetts 02142, USA.,Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, USA.,Howard Hughes Medical Institute, Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Peter S Hammerman
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Mohamed E Abazeed
- Department of Translational Hematology Oncology Research, Cleveland Clinic, 9500 Euclid Avenue/R40, Cleveland, Ohio 44195, USA.,Department of Radiation Oncology, Cleveland Clinic, 9500 Euclid Avenue/T2, Cleveland, Ohio 44195, USA
| |
Collapse
|
516
|
Chen B, Butte AJ. Leveraging big data to transform target selection and drug discovery. Clin Pharmacol Ther 2016; 99:285-97. [PMID: 26659699 PMCID: PMC4785018 DOI: 10.1002/cpt.318] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 12/02/2015] [Indexed: 02/06/2023]
Abstract
The advances of genomics, sequencing, and high throughput technologies have led to the creation of large volumes of diverse datasets for drug discovery. Analyzing these datasets to better understand disease and discover new drugs is becoming more common. Recent open data initiatives in basic and clinical research have dramatically increased the types of data available to the public. The past few years have witnessed successful use of big data in many sectors across the whole drug discovery pipeline. In this review, we will highlight the state of the art in leveraging big data to identify new targets, drug indications, and drug response biomarkers in this era of precision medicine.
Collapse
Affiliation(s)
- B Chen
- Institute for Computational Health Sciences, University of California, San Francisco, San Francisco, California, USA
| | - A J Butte
- Institute for Computational Health Sciences, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
517
|
Wagner BK, Schreiber SL. The Power of Sophisticated Phenotypic Screening and Modern Mechanism-of-Action Methods. Cell Chem Biol 2016; 23:3-9. [PMID: 26933731 PMCID: PMC4779180 DOI: 10.1016/j.chembiol.2015.11.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 11/19/2015] [Accepted: 11/19/2015] [Indexed: 12/14/2022]
Abstract
The enthusiasm for phenotypic screening as an approach for small-molecule discovery has increased dramatically over the last several years. The recent increase in phenotype-based discoveries is in part due to advancements in phenotypic readouts in improved disease models that recapitulate clinically relevant biology in cell culture. Of course, a major historical barrier to using phenotypic assays in chemical biology has been the challenge in determining the mechanism of action (MoA) for compounds of interest. With the combination of medically inspired phenotypic screening and the development of modern MoA methods, we can now start implementing this approach in chemical probe and drug discovery. In this Perspective, we highlight recent advances in phenotypic readouts and MoA determination by discussing several case studies in which both activities were required for understanding the chemical biology involved and, in some cases, advancing toward clinical development.
Collapse
Affiliation(s)
- Bridget K Wagner
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA 02142, USA.
| | - Stuart L Schreiber
- Center for the Science of Therapeutics, Broad Institute, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
518
|
Disease signatures for schizophrenia and bipolar disorder using patient-derived induced pluripotent stem cells. Mol Cell Neurosci 2016; 73:96-103. [PMID: 26777134 DOI: 10.1016/j.mcn.2016.01.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/05/2016] [Accepted: 01/11/2016] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia and bipolar disorder are complex psychiatric disorders that present unique challenges in the study of disease biology. There are no objective biological phenotypes for these disorders, which are characterized by complex genetics and prominent roles for gene-environment interactions. The study of the neurobiology underlying these severe psychiatric disorders has been hindered by the lack of access to the tissue of interest - neurons from patients. The advent of reprogramming methods that enable generation of induced pluripotent stem cells (iPSCs) from patient fibroblasts and peripheral blood mononuclear cells has opened possibilities for new approaches to study relevant disease biology using iPSC-derived neurons. While early studies with patient iPSCs have led to promising and intriguing leads, significant hurdles remain in our attempts to capture the complexity of these disorders in vitro. We present here an overview of studies to date of schizophrenia and bipolar disorder using iPSC-derived neuronal cells and discuss potential future directions that can result in the identification of robust and valid cellular phenotypes that in turn can lay the groundwork for meaningful clinical advances.
Collapse
|
519
|
Antolin AA, Workman P, Mestres J, Al-Lazikani B. Polypharmacology in Precision Oncology: Current Applications and Future Prospects. Curr Pharm Des 2016; 22:6935-6945. [PMID: 27669965 PMCID: PMC5403974 DOI: 10.2174/1381612822666160923115828] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/19/2016] [Indexed: 02/08/2023]
Abstract
Over the past decade, a more comprehensive, large-scale approach to studying cancer genetics and biology has revealed the challenges of tumor heterogeneity, adaption, evolution and drug resistance, while systems-based pharmacology and chemical biology strategies have uncovered a much more complex interaction between drugs and the human proteome than was previously anticipated. In this mini-review we assess the progress and potential of drug polypharmacology in biomarker-driven precision oncology. Polypharmacology not only provides great opportunities for drug repurposing to exploit off-target effects in a new single-target indication but through simultaneous blockade of multiple targets or pathways offers exciting opportunities to slow, overcome or even prevent inherent or adaptive drug resistance. We highlight the many challenges associated with exploiting known or desired polypharmacology in drug design and development, and assess computational and experimental methods to uncover unknown polypharmacology. A comprehensive understanding of the intricate links between polypharmacology, efficacy and safety is urgently needed if we are to tackle the enduring challenge of cancer drug resistance and to fully exploit polypharmacology for the ultimate benefit of cancer patients.
Collapse
Affiliation(s)
- Albert A. Antolin
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
- Systems Pharmacology, Research Program on Biomedical Informatics (GRIB), IMIM Hospital del Mar Medical Research Institute and University Pompeu Fabra, Parc de Recerca Biomèdica, Barcelona, Catalonia, Spain
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Jordi Mestres
- Systems Pharmacology, Research Program on Biomedical Informatics (GRIB), IMIM Hospital del Mar Medical Research Institute and University Pompeu Fabra, Parc de Recerca Biomèdica, Barcelona, Catalonia, Spain
| | - Bissan Al-Lazikani
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| |
Collapse
|