Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Perspective
  • Published:

Pathway and network analysis of cancer genomes

Abstract

Genomic information on tumors from 50 cancer types cataloged by the International Cancer Genome Consortium (ICGC) shows that only a few well-studied driver genes are frequently mutated, in contrast to many infrequently mutated genes that may also contribute to tumor biology. Hence there has been large interest in developing pathway and network analysis methods that group genes and illuminate the processes involved. We provide an overview of these analysis techniques and show where they guide mechanistic and translational investigations.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Major approaches to pathway and network analysis of cancer data.
Figure 2: Pathway and network representations of EGF signaling.

Similar content being viewed by others

References

  1. Newman, W.G. & Black, G.C. Delivery of a clinical genomics service. Genes (Basel) 5, 1001–1017 (2014).

    Article  CAS  Google Scholar 

  2. Lawrence, M.S. et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505, 495–501 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Biankin, A.V. et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491, 399–405 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Imielinski, M. et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 150, 1107–1120 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Banerji, S. et al. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature 486, 405–409 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008).

  7. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609–615 (2011).

  8. Garraway, L.A. & Lander, E.S. Lessons from the cancer genome. Cell 153, 17–37 (2013). This review discusses the advances and findings in cancer genome sequencing as well as current challenges of the field, including the long 'tail' of infrequently mutated genes and the need for functional validation of cancer mutations.

    Article  CAS  PubMed  Google Scholar 

  9. Zack, T.I. et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 45, 1134–1140 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Mack, S.C. et al. Epigenomic alterations define lethal CIMP-positive ependymomas of infancy. Nature 506, 445–450 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gonzalez-Perez, A. et al. Computational approaches to identify functional genetic variants in cancer genomes. Nat. Methods 10, 723–729 (2013). This review from the ICGC-MUCOPA working group discusses methods and recommendations to distinguish functional cancer mutations and to predict cancer driver genes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Leiserson, M.D.M., Blokh, D., Sharan, R. & Raphael, B.J. Simultaneous identification of multiple driver pathways in cancer. PLoS Comput. Biol. 9, e1003054 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Pe'er, D. & Hacohen, N. Principles and strategies for developing network models in cancer. Cell 144, 864–873 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Califano, A., Butte, A.J., Friend, S., Ideker, T. & Schadt, E. Leveraging models of cell regulation and GWAS data in integrative network-based association studies. Nat. Genet. 44, 841–847 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chi, Y.Y., Gribbin, M.J., Johnson, J.L. & Muller, K.E. Power calculation for overall hypothesis testing with high-dimensional commensurate outcomes. Stat. Med. 33, 812–827 (2014).

    Article  PubMed  Google Scholar 

  16. Akavia, U.D. et al. An integrated approach to uncover drivers of cancer. Cell 143, 1005–1017 (2010). This is one of the first studies to integrate molecular data at different network levels to pinpoint tumor dependencies.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Danussi, C. et al. RHPN2 drives mesenchymal transformation in malignant glioma by triggering RhoA activation. Cancer Res. 73, 5140–5150 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hoadley, K.A. et al. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell 158, 929–944 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sonabend, A.M. et al. The transcriptional regulatory network of proneural glioma determines the genetic alterations selected during tumor progression. Cancer Res. 74, 1440–1451 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Carro, M.S. et al. The transcriptional network for mesenchymal transformation of brain tumours. Nature 463, 318–325 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. International Cancer Genome Consortium. International network of cancer genome projects. Nature 464, 993–998 (2010).

  22. Bader, G.D., Cary, M.P. & Sander, C. Pathguide: a pathway resource list. Nucleic Acids Res. 34, D504–D506 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. The Gene Ontology Consortium. The Gene Ontology in 2010: extensions and refinements. Nucleic Acids Res. 38, D331–D335 (2010).

  24. Huang, D.W. et al. DAVID Bioinformatics Resources: expanded annotation database and novel algorithms to better extract biology from large gene lists. Nucleic Acids Res. 35, W169–W175 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Reimand, J., Kull, M., Peterson, H., Hansen, J. & Vilo, J. g:Profiler—a web-based toolset for functional profiling of gene lists from large-scale experiments. Nucleic Acids Res. 35, W193–W200 (2007). g:Profiler is a frequently updated web server for conducting fixed–gene set enrichment analysis of plain and ranked gene lists.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gundem, G. & Lopez-Bigas, N. Sample-level enrichment analysis unravels shared stress phenotypes among multiple cancer types. Genome Med. 4, 28 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Merico, D., Isserlin, R., Stueker, O., Emili, A. & Bader, G.D. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS ONE 5, e13984 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cline, M.S. et al. Integration of biological networks and gene expression data using Cytoscape. Nat. Protoc. 2, 2366–2382 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kanehisa, M., Goto, S., Sato, Y., Furumichi, M. & Tanabe, M. KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Res. 40, D109–D114 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Croft, D. et al. Reactome: a database of reactions, pathways and biological processes. Nucleic Acids Res. 39, D691–D697 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Caspi, R. et al. The MetaCyc database of metabolic pathways and enzymes and the BioCyc collection of Pathway/Genome Databases. Nucleic Acids Res. 42, D459–D471 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Wu, D. & Smyth, G.K. Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res. 40, e133 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Razick, S., Magklaras, G. & Donaldson, I.M. iRefIndex: a consolidated protein interaction database with provenance. BMC Bioinformatics 9, 405 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chatr-Aryamontri, A. et al. The BioGRID interaction database: 2013 update. Nucleic Acids Res. 41, D816–D823 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Kerrien, S. et al. The IntAct molecular interaction database in 2012. Nucleic Acids Res. 40, D841–D846 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. Franceschini, A. et al. STRING v9.1: protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 41, D808–D815 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Warde-Farley, D. et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 38, W214–W220 (2010). GeneMANIA is a web server for integrative analysis of gene lists in the context of molecular interaction networks.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wu, G., Dawson, E., Duong, A., Haw, R. & Stein, L. ReactomeFIViz: a Cytoscape app for pathway and network-based data analysis. F1000Res. 3, 146 (2014). ReactomeFIViz is a Cytoscape app with multiple algorithms for network-based clustering and analysis of the Reactome functional interaction network.

    PubMed  PubMed Central  Google Scholar 

  41. Lan, A. et al. ResponseNet: revealing signaling and regulatory networks linking genetic and transcriptomic screening data. Nucleic Acids Res. 39, W424–W429 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Cerami, E., Demir, E., Schultz, N., Taylor, B.S. & Sander, C. Automated network analysis identifies core pathways in glioblastoma. PLoS ONE 5, e8918 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ciriello, G., Cera, E., Sander, C. & Schultz, N. Mutual exclusivity analysis identifies oncogenic network modules. Genome Res. 22, 398–406 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Glaab, E., Baudot, A., Krasnogor, N., Schneider, R. & Valencia, A. EnrichNet: network-based gene set enrichment analysis. Bioinformatics 28, i451–i457 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wu, G. & Stein, L. A network module-based method for identifying cancer prognostic signatures. Genome Biol. 13, R112 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Chuang, H.Y., Lee, E., Liu, Y.T., Lee, D. & Ideker, T. Network-based classification of breast cancer metastasis. Mol. Syst. Biol. 3, 140 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Leung, A., Bader, G.D. & Reimand, J. HyperModules: identifying clinically and phenotypically significant network modules with disease mutations for biomarker discovery. Bioinformatics 30, 2230–2232 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Reimand, J. & Bader, G.D. Systematic analysis of somatic mutations in phosphorylation signaling predicts novel cancer drivers. Mol. Syst. Biol. 9, 637 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Krallinger, M. et al. The Protein-Protein Interaction tasks of BioCreative III: classification/ranking of articles and linking bio-ontology concepts to full text. BMC Bioinformatics 12, S3 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Kwong, L.N. et al. Oncogenic NRAS signaling differentially regulates survival and proliferation in melanoma. Nat. Med. 18, 1503–1510 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Schadt, E.E. et al. An integrative genomics approach to infer causal associations between gene expression and disease. Nat. Genet. 37, 710–717 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Aytes, A. et al. Cross-species analysis of genome-wide regulatory networks identifies a synergistic interaction between FOXM1 and CENPF that drives prostate cancer malignancy. Cancer Cell 25, 638–651 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Piovan, E. et al. Direct reversal of glucocorticoid resistance by AKT inhibition in acute lymphoblastic leukemia. Cancer Cell 24, 766–776 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. Bandyopadhyay, S. et al. A human MAP kinase interactome. Nat. Methods 7, 801–805 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Vandin, F., Upfal, E. & Raphael, B.J. Algorithms for detecting significantly mutated pathways in cancer. J. Comput. Biol. 18, 507–522 (2011). The HotNet algorithm uses a heat-diffusion model to analyze molecular interaction networks and detect significantly mutated modules in cancer.

    Article  CAS  PubMed  Google Scholar 

  56. Paull, E.O. et al. Discovering causal pathways linking genomic events to transcriptional states using Tied Diffusion Through Interacting Events (TieDIE). Bioinformatics 29, 2757–2764 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Drier, Y., Sheffer, M. & Domany, E. Pathway-based personalized analysis of cancer. Proc. Natl. Acad. Sci. USA 110, 6388–6393 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Tarca, A.L. et al. A novel signaling pathway impact analysis. Bioinformatics 25, 75–82 (2009).

    Article  CAS  PubMed  Google Scholar 

  59. Margolin, A.A. et al. ARACNE: an algorithm for the reconstruction of gene regulatory networks in a mammalian cellular context. BMC Bioinformatics 7 (suppl. 1), S7 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Morris, M.K., Saez-Rodriguez, J., Sorger, P.K. & Lauffenburger, D.A. Logic-based models for the analysis of cell signaling networks. Biochemistry 49, 3216–3224 (2010).

    Article  CAS  PubMed  Google Scholar 

  61. Morris, M.K., Saez-Rodriguez, J., Clarke, D.C., Sorger, P.K. & Lauffenburger, D.A. Training signaling pathway maps to biochemical data with constrained fuzzy logic: quantitative analysis of liver cell responses to inflammatory stimuli. PLoS Comput. Biol. 7, e1001099 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Janes, K.A. et al. A systems model of signaling identifies a molecular basis set for cytokine-induced apoptosis. Science 310, 1646–1653 (2005).

    Article  CAS  PubMed  Google Scholar 

  63. Lee, M.J. et al. Sequential application of anticancer drugs enhances cell death by rewiring apoptotic signaling networks. Cell 149, 780–794 (2012). This study utilized integrative network analysis to identify key rewiring cellular events that informed a combination-based therapeutic strategy for specific tumors.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Saez-Rodriguez, J. et al. Flexible informatics for linking experimental data to mathematical models via DataRail. Bioinformatics 24, 840–847 (2008).

    Article  CAS  PubMed  Google Scholar 

  65. Greenblum, S.I., Efroni, S., Schaefer, C.F. & Buetow, K.H. The PathOlogist: an automated tool for pathway-centric analysis. BMC Bioinformatics 12, 133 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Brubaker, D. et al. Drug Intervention Response Predictions with PARADIGM (DIRPP) identifies drug resistant cancer cell lines and pathway mechanisms of resistance. Pac. Symp. Biocomput. doi:10.1142/9789814583220_0013 (2014).

  67. Vaske, C.J. et al. Inference of patient-specific pathway activities from multi-dimensional cancer genomics data using PARADIGM. Bioinformatics 26, i237–i245 (2010). The PARADIGM algorithm predicts the impact of oncogenic alterations on downstream pathway and network activity by modeling the 'central dogma' of gene expression.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ng, S. et al. PARADIGM-SHIFT predicts the function of mutations in multiple cancers using pathway impact analysis. Bioinformatics 28, i640–i646 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Hill, S.M. et al. Bayesian inference of signaling network topology in a cancer cell line. Bioinformatics 28, 2804–2810 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Sanghvi, J.C. et al. Accelerated discovery via a whole-cell model. Nat. Methods 10, 1192–1195 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Dittrich, M.T., Klau, G.W., Rosenwald, A., Dandekar, T. & Müller, T. Identifying functional modules in protein-protein interaction networks: an integrated exact approach. Bioinformatics 24, i223–i231 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Wu, M., Pastor-Pareja, J.C. & Xu, T. Interaction between RasV12 and scribbled clones induces tumour growth and invasion. Nature 463, 545–548 (2010). This paper demonstrated the importance of cooperation between mutations in cancer in the RAS signaling pathway.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Berry, D.A. Adaptive clinical trials: the promise and the caution. J. Clin. Oncol. 29, 606–609 (2011).

    Article  PubMed  Google Scholar 

  74. Green, R.C. et al. ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing. Genet. Med. 15, 565–574 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge the assistance of J. Jennings during preparation of this manuscript. J.M.S. acknowledges support from the US National Cancer Institute (R01-CA180778 and U24-CA143858), Stand Up To Cancer, the Prostate Cancer Foundation and the Movember Foundation. P.C. is currently funded by a Ludwig Fund Postdoctoral Fellowship. P.C.B. and L.D.S. were supported by the Ontario Institute for Cancer Research through funding provided by the Government of Ontario. P.C.B. was also supported by a Terry Fox Research Institute New Investigator Award and a Canadian Institutes of Health Research New Investigator Award. L.D.S. and G.W. acknowledge support from the US National Institutes of Health (NIH) and National Human Genome Research Institute (P41 HG003751). G.D.B. is supported by NRNB (NIH, National Institute of General Medical Sciences grant number P41 GM103504).

Author information

Authors and Affiliations

Consortia

Corresponding author

Correspondence to Lincoln D Stein.

Ethics declarations

Competing interests

The author declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1–3 (PDF 283 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

the Mutation Consequences and Pathway Analysis working group of the International Cancer Genome Consortium. Pathway and network analysis of cancer genomes. Nat Methods 12, 615–621 (2015). https://doi.org/10.1038/nmeth.3440

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.3440

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer