Nonlinear mixed effects dose response modeling in high throughput drug screens: application to melanoma cell line analysis

Cancer cell lines are often used in high throughput drug screens (HTS) to explore the relationship between cell line characteristics and responsiveness to different therapies. Many current analysis methods infer relationships by focusing on one aspect of cell line drug-specific dose-response curves (DRCs), the concentration causing 50% inhibition of a phenotypic endpoint (IC50). Such methods may overlook DRC features and do not simultaneously leverage information about drug response patterns across cell lines, potentially increasing false positive and negative rates in drug response associations. We consider the application of two methods, each rooted in nonlinear mixed effects (NLME) models, that test the relationship relationships between estimated cell line DRCs and factors that might mitigate response. Both methods leverage estimation and testing techniques that consider the simultaneous analysis of different cell lines to draw inferences about any one cell line. One of the methods is designed to provide an omnibus test of the differences between cell line DRCs that is not focused on any one aspect of the DRC (such as the IC50 value). We simulated different settings and compared the different methods on the simulated data. We also compared the proposed methods against traditional IC50-based methods using 40 melanoma cell lines whose transcriptomes, proteomes, and, importantly, BRAF and related mutation profiles were available. Ultimately, we find that the NLME-based methods are more robust, powerful and, for the omnibus test, more flexible, than traditional methods. Their application to the melanoma cell lines reveals insights into factors that may be clinically useful.

[1]  Trevor Hastie,et al.  Computer Age Statistical Inference: Algorithms, Evidence, and Data Science , 2016 .

[2]  Chad C. Brown,et al.  An adaptive permutation approach for genome-wide association study: evaluation and recommendations for use , 2014, BioData Mining.

[3]  Sridhar Ramaswamy,et al.  Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells , 2012, Nucleic Acids Res..

[4]  T. Speed,et al.  Summaries of Affymetrix GeneChip probe level data. , 2003, Nucleic acids research.

[5]  Alana L. Welm,et al.  Human Primary Tumorgraft Models : Comparisons with Traditional Oncology Pre-Clinical Models and The Clinical Relevance and Utility of Primary Tumorgrafts in Basic and Translational Oncology Research , 2013 .

[6]  R. Shoemaker The NCI60 human tumour cell line anticancer drug screen , 2006, Nature Reviews Cancer.

[7]  Michael P. Morrissey,et al.  Pharmacogenomic agreement between two cancer cell line data sets , 2015, Nature.

[8]  Erik Willems,et al.  Induced Pluripotent Stem Cells in Cardiovascular Drug Discovery , 2013, Circulation research.

[9]  Joshua M. Stuart,et al.  The Cancer Genome Atlas Pan-Cancer analysis project , 2013, Nature Genetics.

[10]  D. Bates,et al.  Fitting Linear Mixed-Effects Models Using lme4 , 2014, 1406.5823.

[11]  Darren Finlay,et al.  Analysis of variability in high throughput screening data: applications to melanoma cell lines and drug responses , 2017, Oncotarget.

[12]  Jeff Shrager,et al.  Rapid learning for precision oncology , 2014, Nature Reviews Clinical Oncology.

[13]  Yongcui Wang,et al.  Inferences of drug responses in cancer cells from cancer genomic features and compound chemical and therapeutic properties , 2016, Scientific Reports.

[14]  Benjamin Haibe-Kains,et al.  Inconsistency in large pharmacogenomic studies , 2013, Nature.

[15]  Adam A. Margolin,et al.  The Cancer Cell Line Encyclopedia enables predictive modeling of anticancer drug sensitivity , 2012, Nature.

[16]  Levi A Garraway,et al.  Precision oncology: an overview. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[17]  Yuri Nikolsky,et al.  Genomic characterization of explant tumorgraft models derived from fresh patient tumor tissue , 2012, Journal of Translational Medicine.

[18]  N. Schork Personalized medicine: Time for one-person trials , 2015, Nature.

[19]  Chris Sander,et al.  Spatial Normalization of Reverse Phase Protein Array Data , 2014, PloS one.

[20]  Ramakant Sharma,et al.  Phylogeny Estimation and Hypothesis Testing using Maximum Likelihood , 2003 .

[21]  Jean-Pierre Gillet,et al.  The clinical relevance of cancer cell lines. , 2013, Journal of the National Cancer Institute.

[22]  P Hougaard,et al.  Confidence bounds for nonlinear dose-response relationships. , 2015, Statistics in medicine.