Multiplexed single-cell profiling of post-perturbation transcriptional responses to define cancer vulnerabilities and therapeutic mechanism of action

Assays to study cancer cell responses to pharmacologic or genetic perturbations are typically restricted to using simple phenotypic readouts such as proliferation rate or the expression of a marker gene. Information-rich assays, such as gene-expression profiling, are generally not amenable to efficient profiling of a given perturbation across multiple cellular contexts. Here, we developed MIX-Seq, a method for multiplexed transcriptional profiling of post-perturbation responses across a mixture of samples with single-cell resolution, using SNP-based computational demultiplexing of single-cell RNA-sequencing data. We show that MIX-Seq can be used to profile responses to chemical or genetic perturbations across pools of 100 or more cancer cell lines, and combine it with Cell Hashing to further multiplex additional experimental conditions, such as multiple post-treatment time points or drug doses. Analyzing the high-content readout of scRNA-seq reveals both shared and context-specific transcriptional response components that can identify drug mechanism of action and can be used to predict long-term cell viability from short-term transcriptional responses to treatment.

[1]  Jonathan M Lee,et al.  Binding of Elongation Factor eEF1A2 to Phosphatidylinositol 4-Kinase β Stimulates Lipid Kinase Activity and Phosphatidylinositol 4-Phosphate Generation* , 2007, Journal of Biological Chemistry.

[2]  Antoine de Weck,et al.  Project DRIVE: A Compendium of Cancer Dependencies and Synthetic Lethal Relationships Uncovered by Large-Scale, Deep RNAi Screening , 2017, Cell.

[3]  Thomas M. Norman,et al.  Perturb-Seq: Dissecting Molecular Circuits with Scalable Single-Cell RNA Profiling of Pooled Genetic Screens , 2016, Cell.

[4]  S. Ramaswamy,et al.  Systematic identification of genomic markers of drug sensitivity in cancer cells , 2012, Nature.

[5]  Trevor Hastie,et al.  Regularization Paths for Generalized Linear Models via Coordinate Descent. , 2010, Journal of statistical software.

[6]  Marilisa Neri,et al.  DRUG-seq for miniaturized high-throughput transcriptome profiling in drug discovery , 2018, Nature Communications.

[7]  Thomas M. Norman,et al.  A Multiplexed Single-Cell CRISPR Screening Platform Enables Systematic Dissection of the Unfolded Protein Response , 2016, Cell.

[8]  J. Boehm,et al.  From cell lines to living biosensors: new opportunities to prioritize cancer dependencies using ex vivo tumor cultures. , 2019, Current opinion in genetics & development.

[9]  Grace X. Y. Zheng,et al.  Massively parallel digital transcriptional profiling of single cells , 2016, Nature Communications.

[10]  Jill P. Mesirov,et al.  Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway , 2017, Nature.

[11]  Defining a Cancer Dependency Map , 2017, Cell.

[12]  Ann E. Sizemore,et al.  Computational correction of copy-number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells , 2017, Nature Genetics.

[13]  Paul A Clemons,et al.  The Connectivity Map: Using Gene-Expression Signatures to Connect Small Molecules, Genes, and Disease , 2006, Science.

[14]  Orna Elroy-Stein,et al.  Regulation of mRNA Translation during cellular division , 2008, Cell cycle.

[15]  Xinmin Cao,et al.  Stress-induced immediate-early gene, egr-1, involves activation of p38/JNK1 , 1998, Oncogene.

[16]  Leland McInnes,et al.  UMAP: Uniform Manifold Approximation and Projection for Dimension Reduction , 2018, ArXiv.

[17]  James M. McFarland,et al.  Pan-cancer single cell RNA-seq uncovers recurring programs of cellular heterogeneity , 2019 .

[18]  Emanuel J. V. Gonçalves,et al.  Prioritization of cancer therapeutic targets using CRISPR–Cas9 screens , 2019, Nature.

[19]  J. Marioni,et al.  Overcoming confounding plate effects in differential expression analyses of single-cell RNA-seq data , 2016, bioRxiv.

[20]  Emanuel J. V. Gonçalves,et al.  A Landscape of Pharmacogenomic Interactions in Cancer , 2016, Cell.

[21]  Angela N. Brooks,et al.  A Next Generation Connectivity Map: L1000 Platform and the First 1,000,000 Profiles , 2017, Cell.

[22]  Paul Hoffman,et al.  Integrating single-cell transcriptomic data across different conditions, technologies, and species , 2018, Nature Biotechnology.

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

[24]  P. Nghiem,et al.  Faculty Opinions recommendation of A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. , 2019, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.

[25]  Gabor T. Marth,et al.  Haplotype-based variant detection from short-read sequencing , 2012, 1207.3907.

[26]  T. Golub,et al.  A Mechanism of Cyclin D1 Action Encoded in the Patterns of Gene Expression in Human Cancer , 2003, Cell.

[27]  Matthew E. Ritchie,et al.  limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.

[28]  Aaron T. L. Lun,et al.  It's DE-licious: A Recipe for Differential Expression Analyses of RNA-seq Experiments Using Quasi-Likelihood Methods in edgeR , 2016, Statistical Genomics.

[29]  Leland McInnes,et al.  UMAP: Uniform Manifold Approximation and Projection , 2018, J. Open Source Softw..

[30]  J. Dempsey,et al.  LY2606368 Causes Replication Catastrophe and Antitumor Effects through CHK1-Dependent Mechanisms , 2015, Molecular Cancer Therapeutics.

[31]  L. Sequist,et al.  Primary Patient-Derived Cancer Cells and Their Potential for Personalized Cancer Patient Care , 2017, Cell reports.

[32]  Duhee Bang,et al.  Multiplexed single-cell RNA-seq via transient barcoding for simultaneous expression profiling of various drug perturbations , 2019, Science Advances.

[33]  F. Bertucci,et al.  Breast cancer cell lines contain functional cancer stem cells with metastatic capacity and a distinct molecular signature. , 2009, Cancer research.

[34]  R. Satija,et al.  Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression , 2019, Genome Biology.

[35]  Duan Shi-lin,et al.  Trametinib, a novel MEK kinase inhibitor, suppresses lipopolysaccharide-induced tumor necrosis factor (TNF)-α production and endotoxin shock. , 2015, Biochemical and biophysical research communications.

[36]  Anne-Laure Boulesteix,et al.  A computationally fast variable importance test for random forests for high-dimensional data , 2015, Adv. Data Anal. Classif..

[37]  Andrea Califano,et al.  PLATE-Seq for genome-wide regulatory network analysis of high-throughput screens , 2017, Nature Communications.

[38]  Chun Jimmie Ye,et al.  Multiplexed droplet single-cell RNA-sequencing using natural genetic variation , 2017, Nature Biotechnology.

[39]  Bertrand Z. Yeung,et al.  Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics , 2018, Genome Biology.

[40]  Allon M. Klein,et al.  Droplet Barcoding for Single-Cell Transcriptomics Applied to Embryonic Stem Cells , 2015, Cell.

[41]  Evan Z. Macosko,et al.  Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets , 2015, Cell.

[42]  S. Pastore,et al.  The MEK Inhibitors Trametinib and Cobimetinib Induce a Type I Interferon Response in Human Keratinocytes , 2017, International journal of molecular sciences.

[43]  L. Coin,et al.  Genotype-free demultiplexing of pooled single-cell RNA-seq , 2019, Genome Biology.

[44]  Luca Scrucca,et al.  mclust 5: Clustering, Classification and Density Estimation Using Gaussian Finite Mixture Models , 2016, R J..

[45]  Thomas M. Norman,et al.  Exploring genetic interaction manifolds constructed from rich phenotypes , 2019, bioRxiv.

[46]  Mark D. Robinson,et al.  Bias, robustness and scalability in differential expression analysis of single-cell RNA-seq data , 2017, bioRxiv.

[47]  Joshua M. Korn,et al.  Next-generation characterization of the Cancer Cell Line Encyclopedia , 2019, Nature.

[48]  Sydney M. Shaffer,et al.  Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance , 2017, Nature.

[49]  Charlotte Soneson,et al.  Bias, robustness and scalability in single-cell differential expression analysis , 2018, Nature Methods.

[50]  Joshua A. Bittker,et al.  Non-oncology drugs are a source of previously unappreciated anti-cancer activity , 2019, bioRxiv.

[51]  Bo Li,et al.  Nuclei multiplexing with barcoded antibodies for single-nucleus genomics , 2018, Nature Communications.

[52]  Oliver Stegle,et al.  Vireo: Bayesian demultiplexing of pooled single-cell RNA-seq data without genotype reference , 2019, Genome Biology.

[53]  L. Vassilev,et al.  In Vivo Activation of the p53 Pathway by Small-Molecule Antagonists of MDM2 , 2004, Science.

[54]  Helena L. Crowell,et al.  On the discovery of subpopulation-specific state transitions from multi-sample multi-condition single-cell RNA sequencing data , 2019, bioRxiv.

[55]  H. Zou,et al.  Regularization and variable selection via the elastic net , 2005 .

[56]  Charles H. Yoon,et al.  Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq , 2016, Science.

[57]  Justin Lamb,et al.  The Connectivity Map: a new tool for biomedical research , 2007, Nature Reviews Cancer.

[58]  T. Golub,et al.  High-throughput identification of genotype-specific cancer vulnerabilities in mixtures of barcoded tumor cell lines , 2016, Nature Biotechnology.

[59]  Yiming Yang,et al.  Cumulus: a cloud-based data analysis framework for large-scale single-cell and single-nucleus RNA-seq , 2019, bioRxiv.

[60]  Joshua C. Gilbert,et al.  An Interactive Resource to Identify Cancer Genetic and Lineage Dependencies Targeted by Small Molecules , 2013, Cell.

[61]  J. Weinstein,et al.  Karyotypic complexity of the NCI-60 drug-screening panel. , 2003, Cancer research.

[62]  Charity W. Law,et al.  voom: precision weights unlock linear model analysis tools for RNA-seq read counts , 2014, Genome Biology.

[63]  Joshua M. Dempster,et al.  Genetic and transcriptional evolution alters cancer cell line drug response , 2018, Nature.

[64]  Mark D. Robinson,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[65]  T. Palomares,et al.  Removal of N‐glycans from cell surface proteins induces apoptosis by reducing intracellular glutathione levels in the rhabdomyosarcoma cell line S4MH , 2000, Biology of the cell.

[66]  Huqun,et al.  Genetic heterogeneity of the epidermal growth factor receptor in non-small cell lung cancer cell lines revealed by a rapid and sensitive detection system, the peptide nucleic acid-locked nucleic acid PCR clamp. , 2005, Cancer research.

[67]  Helga Thorvaldsdóttir,et al.  Molecular signatures database (MSigDB) 3.0 , 2011, Bioinform..

[68]  Y. Benjamini,et al.  Controlling the false discovery rate: a practical and powerful approach to multiple testing , 1995 .