Skip to main content
Top
Published in: BMC Cancer 1/2017

Open Access 01-12-2017 | Research article

Improved anticancer drug response prediction in cell lines using matrix factorization with similarity regularization

Authors: Lin Wang, Xiaozhong Li, Louxin Zhang, Qiang Gao

Published in: BMC Cancer | Issue 1/2017

Login to get access

Abstract

Background

Human cancer cell lines are used in research to study the biology of cancer and to test cancer treatments. Recently there are already some large panels of several hundred human cancer cell lines which are characterized with genomic and pharmacological data. The ability to predict drug responses using these pharmacogenomics data can facilitate the development of precision cancer medicines. Although several methods have been developed to address the drug response prediction, there are many challenges in obtaining accurate prediction.

Methods

Based on the fact that similar cell lines and similar drugs exhibit similar drug responses, we adopted a similarity-regularized matrix factorization (SRMF) method to predict anticancer drug responses of cell lines using chemical structures of drugs and baseline gene expression levels in cell lines. Specifically, chemical structural similarity of drugs and gene expression profile similarity of cell lines were considered as regularization terms, which were incorporated to the drug response matrix factorization model.

Results

We first demonstrated the effectiveness of SRMF using a set of simulation data and compared it with two typical similarity-based methods. Furthermore, we applied it to the Genomics of Drug Sensitivity in Cancer (GDSC) and Cancer Cell Line Encyclopedia (CCLE) datasets, and performance of SRMF exceeds three state-of-the-art methods. We also applied SRMF to estimate the missing drug response values in the GDSC dataset. Even though SRMF does not specifically model mutation information, it could correctly predict drug-cancer gene associations that are consistent with existing data, and identify novel drug-cancer gene associations that are not found in existing data as well. SRMF can also aid in drug repositioning. The newly predicted drug responses of GDSC dataset suggest that mTOR inhibitor rapamycin was sensitive to non-small cell lung cancer (NSCLC), and expression of AK1RC3 and HINT1 may be adjunct markers of cell line sensitivity to rapamycin.

Conclusions

Our analysis showed that the proposed data integration method is able to improve the accuracy of prediction of anticancer drug responses in cell lines, and can identify consistent and novel drug-cancer gene associations compared to existing data as well as aid in drug repositioning.
Appendix
Available only for authorised users
Literature
1.
go back to reference Mirnezami R, Nicholson J, Darzi A. Preparing for precision medicine. N Engl J Med. 2012;366:489–91.CrossRefPubMed Mirnezami R, Nicholson J, Darzi A. Preparing for precision medicine. N Engl J Med. 2012;366:489–91.CrossRefPubMed
2.
go back to reference Xiao G, Ma S, Minna J, Xie Y. Adaptive prediction model in prospective molecular signature-based clinical studies. Clin Cancer Res. 2014;20:531–9.CrossRefPubMed Xiao G, Ma S, Minna J, Xie Y. Adaptive prediction model in prospective molecular signature-based clinical studies. Clin Cancer Res. 2014;20:531–9.CrossRefPubMed
3.
go back to reference Garnett MJ, Edelman EJ, Heidorn SJ, Greenman CD, Dastur A, Lau KW, et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature. 2012;483:570–5.CrossRefPubMedPubMedCentral Garnett MJ, Edelman EJ, Heidorn SJ, Greenman CD, Dastur A, Lau KW, et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature. 2012;483:570–5.CrossRefPubMedPubMedCentral
4.
go back to reference Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–7.CrossRefPubMedPubMedCentral Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–7.CrossRefPubMedPubMedCentral
5.
go back to reference Iorio F, Knijnenburg TA, Vis DJ, Bignell GR, Menden MP, Schubert M, et al. A landscape of Pharmacogenomic interactions in cancer. Cell. 2016;166:740–54.CrossRefPubMedPubMedCentral Iorio F, Knijnenburg TA, Vis DJ, Bignell GR, Menden MP, Schubert M, et al. A landscape of Pharmacogenomic interactions in cancer. Cell. 2016;166:740–54.CrossRefPubMedPubMedCentral
6.
go back to reference Seashore-Ludlow B, Rees MG, Cheah JH, Cokol M, Price EV, Coletti ME, et al. Harnessing connectivity in a large-scale small-molecule sensitivity dataset. Cancer Discov. 2015;5:1210–23.CrossRefPubMedPubMedCentral Seashore-Ludlow B, Rees MG, Cheah JH, Cokol M, Price EV, Coletti ME, et al. Harnessing connectivity in a large-scale small-molecule sensitivity dataset. Cancer Discov. 2015;5:1210–23.CrossRefPubMedPubMedCentral
7.
go back to reference Costello JC, Heiser LM, Georgii E, Gönen M, Menden MP, Wang NJ, et al. A community effort to assess and improve drug sensitivity prediction algorithms. Nat Biotechnol. 2014;32:1202–12.CrossRefPubMedPubMedCentral Costello JC, Heiser LM, Georgii E, Gönen M, Menden MP, Wang NJ, et al. A community effort to assess and improve drug sensitivity prediction algorithms. Nat Biotechnol. 2014;32:1202–12.CrossRefPubMedPubMedCentral
9.
go back to reference Basu A, Bodycombe NE, Cheah JH, Price EV, Liu K, Schaefer GI, et al. An interactive resource to identify cancer genetic and lineage dependencies targeted by small molecules. Cell. 2013;154:1151–61.CrossRefPubMedPubMedCentral Basu A, Bodycombe NE, Cheah JH, Price EV, Liu K, Schaefer GI, et al. An interactive resource to identify cancer genetic and lineage dependencies targeted by small molecules. Cell. 2013;154:1151–61.CrossRefPubMedPubMedCentral
10.
go back to reference Stetson LC, Pearl T, Chen Y, Barnholtz-Sloan JS. Computational identification of multi-omic correlates of anticancer therapeutic response. BMC Genomics. 2014;15:S2.CrossRefPubMedPubMedCentral Stetson LC, Pearl T, Chen Y, Barnholtz-Sloan JS. Computational identification of multi-omic correlates of anticancer therapeutic response. BMC Genomics. 2014;15:S2.CrossRefPubMedPubMedCentral
11.
go back to reference 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
12.
go back to reference Geeleher P, Cox NJ, Huang RS. Clinical drug response can be predicted using baseline gene expression levels and in vitro drug sensitivity in cell lines. Genome Biol. 2014;15:R47.CrossRefPubMedPubMedCentral Geeleher P, Cox NJ, Huang RS. Clinical drug response can be predicted using baseline gene expression levels and in vitro drug sensitivity in cell lines. Genome Biol. 2014;15:R47.CrossRefPubMedPubMedCentral
13.
go back to reference Dong Z, Zhang N, Li C, Wang H, Fang Y, Wang J, et al. Anticancer drug sensitivity prediction in cell lines from baseline gene expression through recursive feature selection. BMC Cancer. 2015;15:489.CrossRefPubMedPubMedCentral Dong Z, Zhang N, Li C, Wang H, Fang Y, Wang J, et al. Anticancer drug sensitivity prediction in cell lines from baseline gene expression through recursive feature selection. BMC Cancer. 2015;15:489.CrossRefPubMedPubMedCentral
14.
15.
go back to reference Menden MP, Iorio F, Garnett M, McDermott U, Benes CH, Ballester PJ, et al. Machine learning prediction of cancer cell sensitivity to drugs based on genomic and chemical properties. PLoS One. 2013;8:e61318.CrossRefPubMedPubMedCentral Menden MP, Iorio F, Garnett M, McDermott U, Benes CH, Ballester PJ, et al. Machine learning prediction of cancer cell sensitivity to drugs based on genomic and chemical properties. PLoS One. 2013;8:e61318.CrossRefPubMedPubMedCentral
16.
go back to reference Ammad-ud-din M, Georgii E, Gönen M, Laitinen T, Kallioniemi O, Wennerberg K, et al. Integrative and personalized QSAR analysis in cancer by Kernelized Bayesian matrix factorization. J Chem Inf Model. 2014;54:2347–59.CrossRefPubMed Ammad-ud-din M, Georgii E, Gönen M, Laitinen T, Kallioniemi O, Wennerberg K, et al. Integrative and personalized QSAR analysis in cancer by Kernelized Bayesian matrix factorization. J Chem Inf Model. 2014;54:2347–59.CrossRefPubMed
17.
go back to reference Zhang N, Wang H, Fang Y, Wang J, Zheng X, Liu XS. Predicting anticancer drug responses using a dual-layer integrated cell line-drug network model. PLoS Comput Biol. 2015;11:e1004498.CrossRefPubMedPubMedCentral Zhang N, Wang H, Fang Y, Wang J, Zheng X, Liu XS. Predicting anticancer drug responses using a dual-layer integrated cell line-drug network model. PLoS Comput Biol. 2015;11:e1004498.CrossRefPubMedPubMedCentral
18.
go back to reference Cortés-Ciriano I, van Westen GJ, Bouvier G, Nilges M, Overington JP, Bender A, et al. Improved large-scale prediction of growth inhibition patterns using the NCI60 cancer cell line panel. Bioinformatics. 2016;32:85–95.PubMed Cortés-Ciriano I, van Westen GJ, Bouvier G, Nilges M, Overington JP, Bender A, et al. Improved large-scale prediction of growth inhibition patterns using the NCI60 cancer cell line panel. Bioinformatics. 2016;32:85–95.PubMed
19.
go back to reference Yap CW. PaDEL-descriptor: an open source software to calculate molecular descriptors and fingerprints. J Comput Chem. 2011;32:1466–74.CrossRefPubMed Yap CW. PaDEL-descriptor: an open source software to calculate molecular descriptors and fingerprints. J Comput Chem. 2011;32:1466–74.CrossRefPubMed
20.
go back to reference Zheng X, Ding H, Mamitsuka H, Zhu S. Collaborative matrix factorization with multiple similarities for predicting drug-target interactions. KDD’13: Proceedings of the 19th ACM SIGKDD International Conference on Knowledge Discovery and Data Mining. 2013;p. 1025–33. Zheng X, Ding H, Mamitsuka H, Zhu S. Collaborative matrix factorization with multiple similarities for predicting drug-target interactions. KDD’13: Proceedings of the 19th ACM SIGKDD International Conference on Knowledge Discovery and Data Mining. 2013;p. 1025–33.
21.
go back to reference Marcotte R, Sayad A, Brown KR, Sanchez-Garcia F, Reimand J, Haider M, et al. Functional genomic landscape of human breast cancer drivers, vulnerabilities, and resistance. Cell. 2016;164:293–309.CrossRefPubMedPubMedCentral Marcotte R, Sayad A, Brown KR, Sanchez-Garcia F, Reimand J, Haider M, et al. Functional genomic landscape of human breast cancer drivers, vulnerabilities, and resistance. Cell. 2016;164:293–309.CrossRefPubMedPubMedCentral
22.
go back to reference Ross DT, Scherf U, Eisen MB, Perou CM, Rees C, Spellman P, et al. Systematic variation in gene expression patterns in human cancer cell lines. Nat Genet. 2000;24:227–35.CrossRefPubMed Ross DT, Scherf U, Eisen MB, Perou CM, Rees C, Spellman P, et al. Systematic variation in gene expression patterns in human cancer cell lines. Nat Genet. 2000;24:227–35.CrossRefPubMed
24.
go back to reference Dupouy S, Doan VK, Wu Z, Mourra N, Liu J, De Wever O, et al. Activation of EGFR, HER2 and HER3 by neurotensin/neurotensin receptor 1 renders breast tumors aggressive yet highly responsive to lapatinib and metformin in mice. Oncotarget. 2014;5:8235–51.CrossRefPubMedPubMedCentral Dupouy S, Doan VK, Wu Z, Mourra N, Liu J, De Wever O, et al. Activation of EGFR, HER2 and HER3 by neurotensin/neurotensin receptor 1 renders breast tumors aggressive yet highly responsive to lapatinib and metformin in mice. Oncotarget. 2014;5:8235–51.CrossRefPubMedPubMedCentral
25.
go back to reference Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang G, Rahmeh M, et al. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66:1630–9.CrossRefPubMed Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang G, Rahmeh M, et al. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66:1630–9.CrossRefPubMed
26.
go back to reference Konecny GE, Winterhoff B, Kolarova T, Qi J, Manivong K, Dering J, et al. Expression of p16 and retinoblastoma determines response to cdk4/6 inhibition in ovarian cancer. Clin Cancer Res. 2011;17:1591–602.CrossRefPubMedPubMedCentral Konecny GE, Winterhoff B, Kolarova T, Qi J, Manivong K, Dering J, et al. Expression of p16 and retinoblastoma determines response to cdk4/6 inhibition in ovarian cancer. Clin Cancer Res. 2011;17:1591–602.CrossRefPubMedPubMedCentral
27.
go back to reference Smolen GA, Sordella R, Muir B, Mohapatra G, Barmettler A, Archibald H, et al. Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752. Proc Natl Acad Sci U S A. 2006;103:2316–21.CrossRefPubMedPubMedCentral Smolen GA, Sordella R, Muir B, Mohapatra G, Barmettler A, Archibald H, et al. Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752. Proc Natl Acad Sci U S A. 2006;103:2316–21.CrossRefPubMedPubMedCentral
28.
go back to reference McDermott U, Sharma SV, Dowell L, Greninger P, Montagut C, Lamb J, et al. Identification of genotype-correlated sensitivity to selective kinase inhibitors by using high-throughput tumor cell line profiling. Proc Natl Acad Sci U S A. 2007;104:19936–41.CrossRefPubMedPubMedCentral McDermott U, Sharma SV, Dowell L, Greninger P, Montagut C, Lamb J, et al. Identification of genotype-correlated sensitivity to selective kinase inhibitors by using high-throughput tumor cell line profiling. Proc Natl Acad Sci U S A. 2007;104:19936–41.CrossRefPubMedPubMedCentral
29.
go back to reference Liang MC, Ma J, Chen L, Kozlowski P, Qin W, Li D, et al. TSC1 loss synergizes with KRAS activation in lung cancer development in the mouse and confers rapamycin sensitivity. Oncogene. 2010;29:1588–97.CrossRefPubMed Liang MC, Ma J, Chen L, Kozlowski P, Qin W, Li D, et al. TSC1 loss synergizes with KRAS activation in lung cancer development in the mouse and confers rapamycin sensitivity. Oncogene. 2010;29:1588–97.CrossRefPubMed
30.
go back to reference Boffa DJ, Luan F, Thomas D, Yang H, Sharma VK, Lagman M, et al. Rapamycin inhibits the growth and metastatic progression of non-small cell lung cancer. Clin Cancer Res. 2004;10:293–300.CrossRefPubMed Boffa DJ, Luan F, Thomas D, Yang H, Sharma VK, Lagman M, et al. Rapamycin inhibits the growth and metastatic progression of non-small cell lung cancer. Clin Cancer Res. 2004;10:293–300.CrossRefPubMed
31.
go back to reference Miller VL, Lin HK, Murugan P, Fan M, Penning TM, Brame LS, et al. Aldo-keto reductase family 1 member C3 (AKR1C3) is expressed in adenocarcinoma and squamous cell carcinoma but not small cell carcinoma. Int J Clin Exp Pathol. 2012;5:278–89.PubMedPubMedCentral Miller VL, Lin HK, Murugan P, Fan M, Penning TM, Brame LS, et al. Aldo-keto reductase family 1 member C3 (AKR1C3) is expressed in adenocarcinoma and squamous cell carcinoma but not small cell carcinoma. Int J Clin Exp Pathol. 2012;5:278–89.PubMedPubMedCentral
32.
go back to reference Yuan BZ, Jefferson AM, Popescu NC, Reynolds SH. Aberrant gene expression in human non small cell lung carcinoma cells exposed to demethylating agent 5-aza-2′-deoxycytidine. Neoplasia. 2014;6:412–9.CrossRef Yuan BZ, Jefferson AM, Popescu NC, Reynolds SH. Aberrant gene expression in human non small cell lung carcinoma cells exposed to demethylating agent 5-aza-2′-deoxycytidine. Neoplasia. 2014;6:412–9.CrossRef
33.
go back to reference Gönen M, Alpaydın E. Multiple kernel learning algorithms. J Mach Learn Res. 2011;12:2211–68. Gönen M, Alpaydın E. Multiple kernel learning algorithms. J Mach Learn Res. 2011;12:2211–68.
34.
go back to reference Wang Y, Jiang R, Wong WH. Modeling the causal regulatory network by integrating chromatin accessibility and transcriptome data. Natl Sci Rev. 2016;3:240–51.CrossRefPubMedPubMedCentral Wang Y, Jiang R, Wong WH. Modeling the causal regulatory network by integrating chromatin accessibility and transcriptome data. Natl Sci Rev. 2016;3:240–51.CrossRefPubMedPubMedCentral
Metadata
Title
Improved anticancer drug response prediction in cell lines using matrix factorization with similarity regularization
Authors
Lin Wang
Xiaozhong Li
Louxin Zhang
Qiang Gao
Publication date
01-12-2017
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2017
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-017-3500-5

Other articles of this Issue 1/2017

BMC Cancer 1/2017 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine