The recurrent architecture of tumour initiation, progression and drug sensitivity

Andrea Califano, Mariano J Alvarez, Andrea Califano, Mariano J Alvarez

Abstract

Recent studies across multiple tumour types are starting to reveal a recurrent regulatory architecture in which genomic alterations cluster upstream of functional master regulator (MR) proteins, the aberrant activity of which is both necessary and sufficient to maintain tumour cell state. These proteins form small, hyperconnected and autoregulated modules (termed tumour checkpoints) that are increasingly emerging as optimal biomarkers and therapeutic targets. Crucially, as their activity is mostly dysregulated in a post-translational manner, rather than by mutations in their corresponding genes or by differential expression, the identification of MR proteins by conventional methods is challenging. In this Opinion article, we discuss novel methods for the systematic analysis of MR proteins and of the modular regulatory architecture they implement, including their use as a valuable reductionist framework to study the genetic heterogeneity of human disease and to drive key translational applications.

Conflict of interest statement

Competing interests statement

The authors declare competing interests: see Web version for details.

Competing interests statement

A. C is founder of DarwinHealth, Inc. M. J. A. has been employed by DarwinHealth, Inc. since March 2016.

Figures

Figure 1. The architecture of tumour checkpoints
Figure 1. The architecture of tumour checkpoints
a | The probability densities of normal and transformed cells are shown in a principal component (PC) projection that captures most of the sample variability of four tumour types: colorectal adenocarcinoma (COAD), kidney renal clear cell carcinoma (KIRC), uterine corpus endometrial cancer (UCEC) and prostate adenocarcinoma (PRAD). These distributions show a clear single-peak structure, suggesting that the regulatory logic of the tumour cell is effective in avoiding occupancy of states that are far away from the mean. Considering that cancer tissue may also be contaminated by extensive lymphocytic and stromal cell infiltration, the variance of the normal and tumour-associated distributions is of quite comparable magnitude. A comprehensive inventory of all tumour types in The Cancer Genome Atlas (TCGA) reveals that only a handful — such as head and neck squamous cell carcinoma (HNSC), kidney renal papillary cell carcinoma (KIRP) and liver hepatocellular carcinoma (LIHC) —present with substantially greater variance than the corresponding normal tissue. b | The proposed regulatory architecture implemented by master regulator (MR) proteins in tumour checkpoints is shown. MRs (blue spheres in shaded area) represent proteins the concerted, aberrant activity of which is both necessary and sufficient for cancer cell state maintenance. Their aberrant activity is induced by genes in their upstream pathways that are mutated in a specific patient (purple spheres) selected from a larger repertoire of candidate driver genes (green spheres), the mutation of which is recurrently detected in large cohorts. Passenger mutations (pale blue spheres) that are not upstream of MRs have no effect on tumour checkpoint activity and thus on the specific phenotype that the checkpoint regulates. Arrows in this diagram show regulatory and signalling interactions, that is, how one gene product regulates other gene products. Black arrows represent crucial top-down interactions leading from patient mutations first to activation of MR proteins in the tumour checkpoint and then to activation of downstream genetic programmes that are required for tumour phenotype presentation. Grey and blue arrows represent additional regulatory interactions that do not affect and are not affected by tumour checkpoint MRs, respectively. Dashed arrows represent feedback loops implemented either between the MR layer and the upstream modulators or between genes regulated by MR proteins and upstream MR modulators. The MR protein module in the shaded area represents the tumour checkpoint. Pink spheres represent genes that are differentially expressed as a result of the aberrant activity of MR proteins in the tumour checkpoint (that is, the tumour gene expression signature). Lightning bolts represent potential therapeutic interventions using pharmacological inhibitors. Inhibiting oncoproteins mutated in a large fraction (for example, 90%)of tumour sub clones will cause relapse owing to the presence of rare, alternative subclones harbouring either alternative or bypass mutations. A bypass mutation is a mutation that activates the pathway downstream of the pharmacological intervention point. By contrast, inhibiting the tumour checkpoint may represent a more effective strategy, as it captures the effect of all upstream mutations.
Figure 2. Dysregulation of homeostatic control following…
Figure 2. Dysregulation of homeostatic control following malignant transformation and activation of dystasis control mechanisms that are responsible for the stability of tumour cell state
This figure shows how normal cell physiology is determined by the energetic landscape of its regulatory networks, enabling cells to follow specific developmental trajectories that are highly insensitive to genetic, epigenetic and environmental variability, thus achieving stable end point states. This process, also known as Waddington canalization, is illustrated in a cartoon showing differentiation from haematopoietic stem cell (HSC), to multi-lymphoid progenitor (MLP) to a fully differentiated human B cell as a set of transitions to states of progressively lower energy and thus higher stability. Disruption of this regulatory landscape by genetic alterations and environmental signals leads to physiological state loss and emergence of novel, stable disease states, for example, diffuse large B cell lymphoma (DLBCL). When multiple, quasi-isoenergetic states emerge, they can lead to coexistence of cells representing distinct tumour phenotypes in the same tumour mass or to tumour cell reprogramming to different states following treatment, a process known as tumour plasticity. For example, it has been shown that cells representing both the mesenchymal and the proneural subtype of glioma can coexist in the same tumour and that a small fraction of cells treated with tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) transiently reprogramme to a TRAIL-resistant state. Whereas normal cell homeostasis presides over the stability of physiological cell states, by making them difficult to escape, we propose that a dysregulated form of these stability control processes (that is, tumour dystasis) is responsible for the stability of tumour-associated cell states and is mechanistically implemented by a small number of master regulator (MR) proteins in a tumour checkpoint.
Figure 3. Diverse genetic alterations in upstream…
Figure 3. Diverse genetic alterations in upstream pathways contribute to aberrant NF-κB activity in DLBCL
Systematic analysis of genes in pathways upstream of the nuclear factor-κB (NF-κB) complex revealed a large repertoire of diffuse large B cell lymphoma (DLBCL)-specific genetic alterations in B cell receptor (BCR) and myeloid differentiation primary response 88 (MYD88) pathways. The presence of these mutations leads to aberrant activation of the canonical p50-RELA heterodimer and associated tumour dependency. These mutations, which are more frequent in the activated B cell (ABC) subtype of DLBCL, have provided the rationale for the clinical development of several BCR pathway inhibitors, such as ibrutinib, a Bruton tyrosine kinase (BTK) inhibitor. CARD11, caspase recruitment domain family member 11; IFN, interferon; IL, interleukin; IRAK, IL-1 receptor-associated kinase; IRF4, interferon regulatory factor 4; ITAM, immune receptor tyrosine-based activation motif; JAK1, Janus kinase 1; MALT1, mucosa-associated lymphoid tissue lymphoma translocation protein 1; PKC, protein kinase C; STAT, signal transducer and activator of transcription; TIR, Toll-interleukin receptor; TRAF6, TNF receptor associated factor 6. Adapted with permission from REF. , Nature Publishing Group.
Figure 4. Protein activity inference from the…
Figure 4. Protein activity inference from the expression of its regulatory targets
a | Protein activity is the ultimate result of a complex cascade of molecular processes, from transcription and translation, to post-translational modification, complex formation and localization to appropriate subcellular compartments. As a result, there are no individual assays that can accurately measure protein activity in proteome-wide fashion. Instead, we have proposed that an accurate estimator of protein activity is represented by the gene expression of its transcriptional targets, that is, its regulon. This rationale is implemented by the Virtual Inference of Protein Activity by Enriched Regulon Analysis (VIPER) algorithm, based on transcription altargets inferred by reverse engineering algorithms such as Algorithm for the Accurate Reconstruction of Cellular Networks (ARACNe). b | When a protein is inactive its targets are randomly distributed in terms of differential expression. c | By contrast, when the same protein is aberrantly activated, its positively regulated targets become significantly overexpressed and its repressed targets become underexpressed. This can be effectively and quantitatively assessed by gene expression enrichment analysis methods. EGFR, epidermal growth factor receptor.
Figure 5. Tumour checkpoint architecture of the…
Figure 5. Tumour checkpoint architecture of the mesenchymal subtype of glioblastoma
Transcription factors involved in the activation of mesenchymal glioblastoma (MES-GBM) subtype are shown in purple. Overall, the six transcription factors shown in this figure—CCAAT/enhancer-binding protein-β (CEBPβ) and CEBPδ are represented by CEBP, for simplicity, as they form homodimers and heterodimers — control 74% of the genes in the mesenchymal signature of high-grade glioma. A region between 2 kb upstream and 2 kb downstream of the transcription start site of the target genes identified by Algorithm for the Accurate Reconstruction of Cellular Networks (ARACNe) was analysed for the presence of putative binding sites. When combined with analysis of gene expression profiles following short hairpin RNA (shRNA)-mediated silencing of these transcription factors, the latter were shown to bind and regulate the large majority of MES-GBM signature genes (shown in pink). In addition, CEBP (both β and δ subunits) and signal transducer and activator of transcription 3 (STAT3) were shown to regulate the other three transcription factors in the tumour checkpoint and to synergistically regulate the state of MES-GBM cells. ACTA2, actin α2; ACTN1, actinin α1; ANGPT2, angiopoietin 2; ANPEP, alanyl aminopeptidase; BACE2, β-site APP-cleaving enzyme 2; B4GALT1, β-1, 4-galactosyltransferase 1; BHLHE40, class E basic helix–loop–helix protein 40; CA12, carbonic anhydrase 12; C1QTNF1, C1q and tumour necrosis factor related protein 1; C1R, complement C1r; C1RL, complement C1r subcomponent like; CHI3L1, chitinase 3 like 1; COL4A2, collagen type IVα1 chain; ECE1, endothelin converting enzyme 1; EFEMP2, EGF containing fibulin like extracellular matrix protein 2; EFNB2, ephrin B2; EHD2, EH domain containing 2; EMP1, epithelial membrane protein 1; ESM1, endothelial cell specific molecule 1; FCGR2A, Fc fragment of IgG receptor IIa; FLNA, filamin A; FOSL2, Fos-related antigen 2; FPRL1, formyl peptide receptor-like 1; HRH1, histamine receptor H1; ICAM1, intercellular adhesion molecule 1; IFITM, interferon induced transmembrane protein; IL32, interleukin-32; TGA7, integrin subunit α7; LIF, leukaemia inhibitory factor; MMP, matrix metalloproteinase; MVP, major vault protein; MYH9, myosin heavy chain 9; MYL9, myosin light chain 9; NRP2, neuropilin 2; OSMR, oncostatin M receptor; PAPPA, pappalysin 1; PDLIM4; PDZ and LIM domain 4; PDPN, podoplanin; PELO, pelota homologue; PI3, peptidase inhibitor 3; PLA2G5, phospholipase A2 group V; PLAU, plasminogen activator, urokinase; PLAUR, PLAU receptor; PVRL2, poliovirus receptor-related 2; PTRF, polymerase I and transcript release factor; RRBP1, ribosome binding protein 1; RUNX1, runt-related transcription factor 1; SGSH, N-sulfoglucosamine sulfohydrolase; S100A11, S100 calcium binding protein A11; SLC39A8, solute carrier family 39 member 8; SOCS3, suppressor of cytokine signalling 3; TAGLN, transgelin; THBD, thrombomodulin; TIMP1, tissue inihibitor of metalloproteinase 1; TMEPAI, transmembrane prostate androgen-induced protein; TNC, tenascin C; TPP1, tripeptidyl peptidase 1; ZYX, zyxin. Adapted with permission from REF. , Nature Publishing Group.

References

    1. Hoadley KA, et al. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell. 2014;158:929–944.
    1. Shah SP, et al. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature. 2012;486:395–399.
    1. Verhaak RG, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17:98–110.
    1. Curtis C, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–352.
    1. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70.
    1. Oakman C, Santarpia L, Di Leo A. Breast cancer assessment tools and optimizing adjuvant therapy. Nat Rev Clin Oncol. 2010;7:725–732.
    1. Chen JC, et al. Identification of causal genetic drivers of human disease through systems-level analysis of regulatory networks. Cell. 2014;159:402–414.
    1. Compagno M, et al. Mutations of multiple genes cause deregulation of NF-κB in diffuse large B-cell lymphoma. Nature. 2009;459:717–721.
    1. Aytes A, et al. Cross-species regulatory network analysis identifies a synergistic interaction between FOXM1 and CENPF that drives prostate cancer malignancy. Cancer Cell. 2014;25:638–651.
    1. Carro MS, et al. The transcriptional network for mesenchymal transformation of brain tumours. Nature. 2010;463:318–325.
    1. Bisikirska B, et al. Elucidation and pharmacological targeting of novel molecular drivers of follicular lymphoma progression. Cancer Res. 2016;76:664–674.
    1. Yang J, et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 2004;117:927–939.
    1. Nambu JR, Lewis JO, Wharton KA, Jr, Crews ST. The Drosophila single-minded gene encodes a helix–loop–helix protein that acts as a master regulator of CNS midline development. Cell. 1991;67:1157–1167.
    1. Resnick MA, Tomso D, Inga A, Menendez D, Bell D. Functional diversity in the gene network controlled by the master regulator p53 in humans. Cell Cycle. 2005;4:1026–1029.
    1. Klapper LN, Kirschbaum MH, Sela M, Yarden Y. Biochemical and clinical implications of the ErbB/HER signaling network of growth factor receptors. Adv Cancer Res. 2000;77:25–79.
    1. Perou CM, et al. Molecular portraits of human breast tumours. Nature. 2000;406:747–752.
    1. Dalla-Favera R, et al. Human c-myc oncogene is located on the region of chromosome 8 that is translocated in Burkitt lymphoma cells. Proc Natl Acad Sci USA. 1982;79:7824–7827.
    1. Klein U, et al. Transcriptional analysis of the B cell germinal center reaction. Proc Natl Acad Sci USA. 2003;100:2639–2644.
    1. Akavia UD, et al. An integrated approach to uncover drivers of cancer. Cell. 2010;143:1005–1017.
    1. Gu J, et al. Gene module based regulator inference identifying miR-139 as a tumor suppressor in colorectal cancer. Mol Biosyst. 2014;10:3249–3254.
    1. Ergun A, Lawrence CA, Kohanski MA, Brennan TA, Collins JJ. A network biology approach to prostate cancer. Mol Syst Biol. 2007;3:82.
    1. Cannon WB. Organization for physiological homeostasis. Physiol Rev. 1929;9:399–431.
    1. Waddington CH. Canalization of development and genetic assimilation of acquired characters. Nature. 1959;183:1654–1655.
    1. Waddington CH. Genetic assimilation. Adv Genet. 1961;10:257–293.
    1. Siegel PM, Massague J. Cytostatic and apoptotic actions of TGF-β in homeostasis and cancer. Nat Rev Cancer. 2003;3:807–821.
    1. Carter SB. Tissue homeostasis and the biological basis of cancer. Nature. 1968;220:970–974.
    1. Young SR, et al. Establishment and serial passage of cell cultures derived from LuCaP xenografts. Prostate. 2013;73:1251–1262.
    1. Barretina J, et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–607.
    1. Rodriguez-Barrueco R, et al. Inhibition of the autocrine IL-6–JAK2–STAT3–calprotectin axis as targeted therapy for HR−/HER2+ breast cancers. Genes Dev. 2015;29:1631–1648.
    1. Piovan E, et al. Direct reversal of glucocorticoid resistance by AKT inhibition in acute lymphoblastic leukemia. Cancer Cell. 2013;24:766–776.
    1. Mitrofanova A, Aytes A, Shen C, Abate-Shen C, Califano A. A systems biology approach to predict drug response for human prostate cancer based on in vivo preclinical analyses of mouse models. Cell Rep. 2015;12:1–12.
    1. Davis RE, Brown KD, Siebenlist U, Staudt LM. Constitutive nuclear factor kappaB activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells. J Exp Med. 2001;194:1861–1874.
    1. Ngo VN, et al. Oncogenically active MYD88 mutations in human lymphoma. Nature. 2011;470:115–119.
    1. Davis RE, et al. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010;463:88–92.
    1. Luo J, Solimini NL, Elledge SJ. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell. 2009;136:823–837.
    1. Wayne DW. Kolmogorov–Smirnov One-sample Test: Applied Nonparametric Statistics. 2. pp. 319–330. (PWS-Kent, 1990)
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–15550.
    1. Alvarez MJ, et al. Functional characterization of somatic mutations in cancer using network-based inference of protein activity. Nat Genet. 2016;48:838–847.
    1. Lefebvre C, et al. A human B-cell interactome identifies MYB and FOXM1 as master regulators of proliferation in germinal centers. Mol Syst Biol. 2010;6:377.
    1. Castro MA, et al. Regulators of genetic risk of breast cancer identified by integrative network analysis. Nat Genet. 2016;48:12–21.
    1. Zhang S, et al. Stroma-associated master regulators of molecular subtypes predict patient prognosis in ovarian cancer. Sci Rep. 2015;5:16066.
    1. Yepes S, Torres MM, Lopez-Kleine L. Regulatory network reconstruction reveals genes with prognostic value for chronic lymphocytic leukemia. BMC Genomics. 2015;16:1002.
    1. Remo A, et al. Systems biology analysis reveals NFAT5 as a novel biomarker and master regulator of inflammatory breast cancer. J Transl Med. 2015;13:138.
    1. Fletcher MN, et al. Master regulators of FGFR2 signalling and breast cancer risk. Nat Commun. 2013;4:2464.
    1. Tanaka H, Ogishima S. Network biology approach to epithelial-mesenchymal transition in cancer metastasis: three stage theory. J Mol Cell Biol. 2015;7:253–266.
    1. Piao G, et al. A computational procedure for identifying master regulator candidates: a case study on diabetes progression in Goto–Kakizaki rats. BMC Syst Biol. 2012;6(Suppl 1):S2.
    1. Liang Y, et al. Transcriptional network analysis identifies BACH1 as a master regulator of breast cancer bone metastasis. J Biol Chem. 2012;287:33533–33544.
    1. Califano A, Butte AJ, Friend S, Ideker T, Schadt E. Leveraging models of cell regulation and GWAS data in integrative network-based association studies. Nat Genet. 2012;44:841–847.
    1. Pe’er D, Hacohen N. Principles and strategies for developing network models in cancer. Cell. 2011;144:864–873.
    1. Tovar H, Garcia-Herrera R, Espinal-Enriquez J, Hernandez-Lemus E. Transcriptional master regulator analysis in breast cancer genetic networks. Comput Biol Chem. 2015;59B:67–77.
    1. Sartor IT, Zeidan-Chulia F, Albanus RD, Dalmolin RJ, Moreira JC. Computational analyses reveal a prognostic impact of TULP3 as a transcriptional master regulator in pancreatic ductal adenocarcinoma. Mol Biosyst. 2014;10:1461–1468.
    1. Lim WK, Lyashenko E, Califano A. Master regulators used as breast cancer metastasis classifiers. Pac Symp Biocomput. 2009;14:504–515.
    1. Ikiz B, et al. Dissecting the regulatory machinery of neurodegeneration in an in vitro model of amyotrophic lateral sclerosis. Cell Rep. 2015;12:335–345.
    1. Aubry S, et al. Assembly and interrogation of Alzheimer’s disease genetic networks reveal novel regulators of progression. PLoS ONE. 2015;10:e0120352.
    1. Brichta L, et al. Identification of neurodegenerative factors using translatome-regulatory network analysis. Nat Neurosci. 2015;18:1325–1333.
    1. Repunte-Canonigo V, et al. Identifying candidate drivers of alcohol dependence-induced excessive drinking by assembly and interrogation of brain-specific regulatory networks. Genome Biol. 2015;16:68.
    1. Kushwaha R, et al. Interrogation of a context-specific transcription factor network identifies novel regulators of pluripotency. Stem Cells. 2015;33:367–377.
    1. Ravasi T, et al. An atlas of combinatorial transcriptional regulation in mouse and man. Cell. 2010;140:744–752.
    1. Basso K, et al. Reverse engineering of regulatory networks in human B cells. Nat Genet. 2005;37:382–390.
    1. ENCODE Project Consortium. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 2007;447:799–816.
    1. Sumazin P, et al. An extensive microRNA-mediated network of RNA–RNA interactions regulates established oncogenic pathways in glioblastoma. Cell. 2011;147:370–381.
    1. Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB. Prediction of mammalian microRNA targets. Cell. 2003;115:787–798.
    1. Friedlander MR, et al. Discovering microRNAs from deep sequencing data using miRDeep. Nat Biotechnol. 2008;26:407–415.
    1. Rual JF, et al. Towards a proteome-scale map of the human protein-protein interaction network. Nature. 2005;437:1173–1178.
    1. Bandyopadhyay S, et al. A human MAP kinase interactome. Nat Methods. 2010;7:801–805.
    1. Wang K, et al. Genome-wide identification of post-translational modulators of transcription factor activity in human B cells. Nat Biotechnol. 2009;27:829–839.
    1. Zhang QC, et al. Structure-based prediction of protein-protein interactions on a genome-wide scale. Nature. 2012;490:556–560.
    1. AlQuraishi M, Koytiger G, Jenney A, MacBeath G, Sorger PK. A multiscale statistical mechanical framework integrates biophysical and genomic data to assemble cancer networks. Nat Genet. 2014;46:1363–1367.
    1. Faith JJ, et al. Large-scale mapping and validation of Escherichia coli transcriptional regulation from a compendium of expression profiles. PLoS Biol. 2007;5:e8.
    1. Friedman N. Inferring cellular networks using probabilistic graphical models. Science. 2004;303:799–805.
    1. Kundaje A, et al. Learning regulatory programs that accurately predict differential expression with MEDUSA. Ann NY Acad Sci. 2007;1115:178–202.
    1. Kramer A, Green J, Pollard J, Jr, Tugendreich S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics. 2014;30:523–530.
    1. Maher B. ENCODE: the human encyclopaedia. Nature. 2012;489:46–48.
    1. Palomero T, et al. NOTCH1 directly regulates c-MYC and activates a feed-forward-loop transcriptional network promoting leukemic cell growth. Proc Natl Acad Sci USA. 2006;103:18261–18266.
    1. Affara M, et al. Vasohibin-1 is identified as a master-regulator of endothelial cell apoptosis using gene network analysis. BMC Genomics. 2013;14:23.
    1. Werner HM, Mills GB, Ram PT. Cancer systems biology: a peek into the future of patient care? Nat Rev Clin Oncol. 2014;11:167–176.
    1. Saito M, et al. BCL6 suppression of BCL2 via Miz1 and its disruption in diffuse large B cell lymphoma. Proc Natl Acad Sci USA. 2009;106:11294–11299.
    1. Luscombe NM, et al. Genomic analysis of regulatory network dynamics reveals large topological changes. Nature. 2004;431:308–312.
    1. Phillips HS, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell. 2006;9:157–173.
    1. Brennan CW, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155:462–477.
    1. Wapinski OL, et al. Hierarchical mechanisms for direct reprogramming of fibroblasts to neurons. Cell. 2013;155:621–635.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674.
    1. Chudnovsky Y, et al. ZFHX4 interacts with the NuRD core member CHD4 and regulates the glioblastoma tumor-initiating cell state. Cell Rep. 2014;6:313–324.
    1. Taylor BS, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010;18:11–22.
    1. US National Library of Medicine. . 2014 .
    1. Tzoneva G, et al. Activating mutations in the NT5C2 nucleotidase gene drive chemotherapy resistance in relapsed ALL. Nat Med. 2013;19:368–371.
    1. Woyach JA, et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014;370:2286–2294.
    1. Flusberg DA, Sorger PK. Modulating cell-to-cell variability and sensitivity to death ligands by co-drugging. Phys Biol. 2013;10:035002.
    1. Fenner A. Prostate cancer: next-generation RNA sequencing identifies gene signature of neuroendocrine differentiation in prostate tumors. Nat Rev Urol. 2012;9:8.
    1. Van Vlierberghe P, Ferrando A. The molecular basis of T cell acute lymphoblastic leukemia. J Clin Invest. 2012;122:3398–3406.
    1. Wei G, et al. Gene expression-based chemical genomics identifies rapamycin as a modulator of MCL1 and glucocorticoid resistance. Cancer Cell. 2006;10:331–342.
    1. Rubin EH, Gilliland DG. Drug development and clinical trials—the path to an approved cancer drug. Nat Rev Clin Oncol. 2012;9:215–222.
    1. Shelanski M, et al. A systems approach to drug discovery in Alzheimer’s disease. Neurotherapeutics. 2015;12:126–131.
    1. Schadt EE, et al. An integrative genomics approach to infer causal associations between gene expression and disease. Nat Genet. 2005;37:710–717.
    1. Hofree M, Shen JP, Carter H, Gross A, Ideker T. Network-based stratification of tumor mutations. Nat Methods. 2013;10:1108–1115.
    1. Alvarez MJ, Chen JC, Califano A. DIGGIT: a Bioconductor package to infer genetic variants driving cellular phenotypes. Bioinformatics. 2015;31:4032–4034.
    1. Weinstein IB. Cancer. Addiction to oncogenes—the Achilles heal of cancer. Science. 2002;297:63–64.
    1. Druker BJ, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med. 1996;2:561–566.
    1. Wang X, Haswell JR, Roberts CW. Molecular pathways: SWI/SNF (BAF) complexes are frequently mutated in cancer—mechanisms and potential therapeutic insights. Clin Cancer Res. 2014;20:21–27.
    1. Vazquez M, Valencia A, Pons T. Structure-PPi: a module for the annotation of cancer-related single-nucleotide variants at protein-protein interfaces. Bioinformatics. 2015;31:2397–2399.
    1. Morris JPt, Wang SC, Hebrok M. KRAS, Hedgehog, Wnt and the twisted developmental biology of pancreatic ductal adenocarcinoma. Nat Rev Cancer. 2010;10:683–695.
    1. Dhomen N, Marais R. BRAF signaling and targeted therapies in melanoma. Hematol Oncol Clin North Am. 2009;23:529–545.
    1. Davoli A, Hocevar BA, Brown TL. Progression and treatment of HER2-positive breast cancer. Cancer Chemother Pharmacol. 2010;65:611–623.
    1. Bansal M, et al. A community computational challenge to predict the activity of pairs of compounds. Nat Biotechnol. 2014;32:1213–1222.
    1. Woo JH, et al. Elucidating compound mechanism of action by network dysregulation analysis in pertubed cells. Cell. 2015;162:441–451.
    1. Tallarida RJ. An overview of drug combination analysis with isobolograms. J Pharmacol Exp Ther. 2006;319:1–7.
    1. Patel AP, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014;344:1396–1401.
    1. Park HS, Jun CH. A simple and fast algorithm for K-medoids clustering. Expert Systems With Appl. 2009;36:3336–3341.
    1. Lefebvre C, Rieckhof G, Califano A. Reverse-engineering human regulatory networks. Wiley Interdiscip Rev Syst Biol Med. 2012;4:311–325.
    1. Linding R, et al. Systematic discovery of in vivo phosphorylation networks. Cell. 2007;129:1415–1426.
    1. Srivas R, et al. Assembling global maps of cellular function through integrative analysis of physical and genetic networks. Nat Protoc. 2011;6:1308–1323.
    1. Rhodes DR, et al. Mining for regulatory programs in the cancer transcriptome. Nat Genet. 2005;37:579–583.
    1. Lachmann A, et al. ChEA: transcription factor regulation inferred from integrating genome-wide ChIP-X experiments. Bioinformatics. 2010;26:2438–2444.
    1. Needham CJ, Bradford JR, Bulpitt AJ, Westhead DR. Inference in Bayesian networks. Nat Biotechnol. 2006;24:51–53.
    1. Yeung MK, Tegner J, Collins JJ. Reverse engineering gene networks using singular value decomposition and robust regression. Proc Natl Acad Sci USA. 2002;99:6163–6168.
    1. Martinez MR, et al. Quantitative modeling of the terminal differentiation of B cells and mechanisms of lymphomagenesis. Proc Natl Acad Sci USA. 2012;109:2672–2677.
    1. Polynikis A, Hogan SJ, di Bernardo M. Comparing different ODE modelling approaches for gene regulatory networks. J Theor Biol. 2009;261:511–530.
    1. Stuart JM, Segal E, Koller D, Kim SK. A gene-coexpression network for global discovery of conserved genetic modules. Science. 2003;302:249–255.
    1. Horvath S, Dong J. Geometric interpretation of gene coexpression network analysis. PLoS Comput Biol. 2008;4:e1000117.
    1. Jansen R, et al. A Bayesian networks approach for predicting protein-protein interactions from genomic data. Science. 2003;302:449–453.
    1. He F, Balling R, Zeng AP. Reverse engineering and verification of gene networks: principles, assumptions, and limitations of present methods and future perspectives. J Biotechnol. 2009;144:190–203.
    1. Stolovitzky G, Prill RJ, Califano A. Lessons from the DREAM2 Challenges. Ann NY Acad Sci. 2009;1158:159–195.
    1. Marbach D, et al. Revealing strengths and weaknesses of methods for gene network inference. Proc Natl Acad Sci USA. 2010;107:6286–6291.
    1. Bussemaker HJ, Li H, Siggia ED. Regulatory element detection using correlation with expression. Nat Genet. 2001;27:167–171.
    1. Fiedler D, et al. Functional organization of the S cerevisiae phosphorylation network. Cell. 2009;136:952–963.
    1. Mani KM, et al. A systems biology approach to prediction of oncogenes and molecular perturbation targets in B-cell lymphomas. Mol Syst Biol. 2008;4:169.
    1. Ideker T, Krogan NJ. Differential network biology. Mol Syst Biol. 2012;8:565.
    1. Barrios-Rodiles M, et al. High-throughput mapping of a dynamic signaling network in mammalian cells. Science. 2005;307:1621–1625.
    1. Saez-Rodriguez J, et al. Comparing signaling networks between normal and transformed hepatocytes using discrete logical models. Cancer Res. 2011;71:5400–5411.
    1. Menashe I, et al. Pathway analysis of breast cancer genome-wide association study highlights three pathways and one canonical signaling cascade. Cancer Res. 2010;70:4453–4459.
    1. Laoukili J, et al. FoxM1 is required for execution of the mitotic programme and chromosome stability. Nature Cell Biol. 2005;7:126–136.
    1. Basu A, et al. An interactive resource to identify cancer genetic and lineage dependencies targeted by small molecules. Cell. 2013;154:1151–1161.
    1. Flusberg DA, Roux J, Spencer SL, Sorger PK. Cells surviving fractional killing by TRAIL exhibit transient but sustainable resistance and inflammatory phenotypes. Mol Biol Cell. 2013;24:2186–2200.
    1. Roschewski M, Staudt LM, Wilson WH. Diffuse large B-cell lymphoma-treatment approaches in the molecular era. Nat Rev Clin Oncol. 2014;11:12–23.

Source: PubMed

3
订阅