Onco-Multi-OMICS Approach: A New Frontier in Cancer Research

Sajib Chakraborty, Md Ismail Hosen, Musaddeque Ahmed, Hossain Uddin Shekhar, Sajib Chakraborty, Md Ismail Hosen, Musaddeque Ahmed, Hossain Uddin Shekhar

Abstract

The acquisition of cancer hallmarks requires molecular alterations at multiple levels including genome, epigenome, transcriptome, proteome, and metabolome. In the past decade, numerous attempts have been made to untangle the molecular mechanisms of carcinogenesis involving single OMICS approaches such as scanning the genome for cancer-specific mutations and identifying altered epigenetic-landscapes within cancer cells or by exploring the differential expression of mRNA and protein through transcriptomics and proteomics techniques, respectively. While these single-level OMICS approaches have contributed towards the identification of cancer-specific mutations, epigenetic alterations, and molecular subtyping of tumors based on gene/protein-expression, they lack the resolving-power to establish the casual relationship between molecular signatures and the phenotypic manifestation of cancer hallmarks. In contrast, the multi-OMICS approaches involving the interrogation of the cancer cells/tissues in multiple dimensions have the potential to uncover the intricate molecular mechanism underlying different phenotypic manifestations of cancer hallmarks such as metastasis and angiogenesis. Moreover, multi-OMICS approaches can be used to dissect the cellular response to chemo- or immunotherapy as well as discover molecular candidates with diagnostic/prognostic value. In this review, we focused on the applications of different multi-OMICS approaches in the field of cancer research and discussed how these approaches are shaping the field of personalized oncomedicine. We have highlighted pioneering studies from "The Cancer Genome Atlas (TCGA)" consortium encompassing integrated OMICS analysis of over 11,000 tumors from 33 most prevalent forms of cancer. Accumulation of huge cancer-specific multi-OMICS data in repositories like TCGA provides a unique opportunity for the systems biology approach to tackle the complexity of cancer cells through the unification of experimental data and computational/mathematical models. In future, systems biology based approach is likely to predict the phenotypic changes of cancer cells upon chemo-/immunotherapy treatment. This review is sought to encourage investigators to bring these different approaches together for interrogating cancer at molecular, cellular, and systems levels.

Figures

Figure 1
Figure 1
Pyramid of complexity. The pyramid represents the flow of information from genome (top) to transcriptome (middle), to proteome (bottom). The complexity increases from genome to proteome (indicated by down arrow). The complexity of transcriptome is largely mediated by temporal dynamics and alternative splicing. In contrast, spatiotemporal dynamics and posttranslational modifications (PTMs) are mainly responsible for high proteome complexity. Examples of PTMs include phosphorylation (P) and acetylation (Ac).
Figure 2
Figure 2
Schematic diagram representing the basic steps of NGS and mass-spectrometry. NGS (left) can be used for both genomic DNA and RNA-sequencing. Mass-spectrometry based proteomics (right) are typically used to identify and quantify large amount of proteins from cells, tissues, and body fluids. 1Primary cells and tissues from patients can be mixed with labelled proteins, typically extracted from cell lines cultured in presence of stable isotopically labelled amino acids. This method is called Super-SILAC. Proteomics can also be done without any labelling steps. This method is known as label free-quantification (LFQ). 2Peptides obtained after tryptic digestion can also be labelled chemically by methods known as “Tandem Mass Tag (TMT)” or “Isobaric tags for relative and absolute quantitation (iTRAQ)”.
Figure 3
Figure 3
Proteomics and phosphoproteomics driven identification of the aberrant regulation of signalling pathways leading to poor patient survival. (a) Aberrant PDGFRB signalling pathway induces angiogenesis in patients and results in poor survival. Phosphorylated and unphosphorylated forms of proteins are indicated by blue and orange color. The directions of arrows indicate the regulation—up and down arrows indicate upregulation and downregulation, respectively. Colors of the arrows indicate the phosphoform or the total protein detected by (phospho)proteomics experiments. Blue and red arrows indicate phospho- and total protein, respectively. (b) Integrin-linked kinase pathway induces cell mobility and invasion, leading to poor patient survival. Two signalling pathways, MAPK (green) and PI3K (purple), are highlighted. Both these pathways were found to be upregulated in cancer patients with poor survival.
Figure 4
Figure 4
Application of NGS and mass-spectrometry (LC-MS/MS) based OMICS techniques in cancer research. Genomics, epigenomics, and transcriptomics are based on NGS techniques; whereas proteomics and metabolomics are driven by mass-spectrometric (LC-MS/MS) technique. The principal application of genomics, epigenomics, and transcriptomics is screening of genome-wide somatic mutations, identification of altered epigenomic landscape, and exploring differential RNA expression, respectively. The major application of proteomics/metabolomics is identification of differentially regulated proteins/phosphoproteins/metabolites. The integration of NGS-based techniques can identify the concordance or discordance between copy number alterations (CNAs), promoter/gene-body methylation, and RNA levels. Integration of NGS and LC-MS/MS based techniques may result in the correlation analysis between CNAs, promoter/gene-body methylation, and mRNA levels with protein/metabolite levels.

References

    1. Hasin Y., Seldin M., Lusis A. Multi-omics approaches to disease. Genome Biology. 2017;18(1)
    1. Manzoni C., Kia D. A., Vandrovcova J., et al. Genome, transcriptome and proteome: the rise of omics data and their integration in biomedical sciences. Briefings in Bioinformatics. 2018;19(2):286–302. doi: 10.1093/bib/bbw114.
    1. Hager G. L., McNally J. G., Misteli T. Transcription Dynamics. Molecular Cell. 2009;35(6):741–753. doi: 10.1016/j.molcel.2009.09.005.
    1. Kandoth C., McLellan M. D., Vandin F., et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;502(7471):333–339. doi: 10.1038/nature12634.
    1. Lawrence M. S., Stojanov P., Polak P. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499:214–218. doi: 10.1038/nature12213.
    1. Piunti A., Shilatifard A. Epigenetic balance of gene expression by polycomb and compass families. Science. 2016;352(6290)
    1. Raj U., Aier I., Semwal R., Varadwaj P. K. Identification of novel dysregulated key genes in Breast cancer through high throughput ChIP-Seq data analysis. Scientific Reports. 2017;7(1)3229
    1. Li J., Ching T., Huang S., Garmire L. X. Using epigenomics data to predict gene expression in lung cancer. BMC Bioinformatics. 2015;16(5):p. S10.
    1. Serre D., Lee B. H., Ting A. H. MBD-isolated Genome Sequencing provides a high-throughput and comprehensive survey of DNA methylation in the human genome. Nucleic Acids Research. 2010;38(2):391–399.
    1. Vidal E., Sayols S., Moran S., et al. A DNA methylation map of human cancer at single base-pair resolution. Oncogene. 2017;36(40):5648–5657. doi: 10.1038/onc.2017.176.
    1. Djebali S., Davis C. A., Merkel A. Landscape of transcription in human cells. Nature. 2012;489:101–108. doi: 10.1038/nature11233.
    1. Li M., Sun Q., Wang X. Transcriptional landscape of human cancers. Oncotarget . 2017;8(21):34534–34551.
    1. Cancer Genome Atlas Research N. The Cancer Genome Atlas Pan-Cancer analysis project. Nature Genetics. 2013;45(10):1113–1120.
    1. Li J., Lu Y., Akbani R., et al. TCPA: A resource for cancer functional proteomics data. Nature Methods. 2013;10(11):1046–1047. doi: 10.1038/nmeth.2650.
    1. Iwamoto N., Shimada T. Recent advances in mass spectrometry-based approaches for proteomics and biologics: Great contribution for developing therapeutic antibodies. Pharmacology & Therapeutics. 2017;185:147–154.
    1. Cox J., Mann M. Maxquant enables high peptide identification rates, individualized ppb-range mass accuracies and proteome-wide protein quantification. Nature Biotechnology. 2008;26(12):1367–1372. doi: 10.1038/nbt.1511.
    1. Kim M.-S., Pinto S. M., Getnet D., et al. A draft map of the human proteome. Nature. 2014;509(7502):575–581. doi: 10.1038/nature13302.
    1. Wilhelm M., Schlegl J., Hahne H., et al. Mass-spectrometry-based draft of the human proteome. Nature. 2014;509(7502):582–587. doi: 10.1038/nature13319.
    1. Shruthi B. S., Vinodhkumar P., Selvamani Proteomics: A new perspective for cancer. Advanced Biomedical Research. 2016;5(67)
    1. Dunn W. B., Broadhurst D. I., Atherton H. J., Goodacre R., Griffin J. L. Systems level studies of mammalian metabolomes: the roles of mass spectrometry and nuclear magnetic resonance spectroscopy. Chemical Society Reviews. 2011;40(1):387–426. doi: 10.1039/b906712b.
    1. Nagana Gowda G. A., Djukovic D. Overview of mass spectrometry-based metabolomics: Opportunities and challenges. Methods in Molecular Biology. 2014;1198:3–12. doi: 10.1007/978-1-4939-1258-2_1.
    1. Hanahan D., Weinberg R. A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Davies M. A., Samuels Y. Analysis of the genome to personalize therapy for melanoma. Oncogene. 2010;29(41):5545–5555. doi: 10.1038/onc.2010.323.
    1. Berdasco M., Esteller M. Aberrant epigenetic landscape in cancer: how cellular identity goes awry. Developmental Cell. 2010;19(5):698–711. doi: 10.1016/j.devcel.2010.10.005.
    1. Karnoub A. E., Weinberg R. A. Chemokine networks and breast cancer metastasis. Breast Disease. 2006;26(1):75–85.
    1. Tyanova S., Albrechtsen R., Kronqvist P., Cox J., Mann M., Geiger T. Proteomic maps of breast cancer subtypes. Nature Communications. 2016;7 doi: 10.1038/ncomms10259.10259
    1. Warburg O. On respiratory impairment in cancer cells. Science. 1956;124(3215):269–270.
    1. Kennedy K. M., Dewhirst M. W. Tumor metabolism of lactate: The influence and therapeutic potential for MCT and CD147 regulation. Future Oncology. 2010;6(1):127–148. doi: 10.2217/fon.09.145.
    1. Hassler M. R., Egger G. Epigenomics of cancer - Emerging new concepts. Biochimie. 2012;94(11):2219–2230. doi: 10.1016/j.biochi.2012.05.007.
    1. Chakravarthi B. V. S. K., Nepal S., Varambally S. Genomic and epigenomic alterations in cancer. The American Journal of Pathology. 2016;186(7):1724–1735. doi: 10.1016/j.ajpath.2016.02.023.
    1. Wilson A. S., Power B. E., Molloy P. L. DNA hypomethylation and human diseases. Biochimica et Biophysica Acta. 2007;1775(1):138–162. doi: 10.1016/j.bbcan.2006.08.007.
    1. Sharma S., Kelly T. K., Jones P. A. Epigenetics in cancer. Carcinogenesis. 2010;31(1):27–36. doi: 10.1093/carcin/bgp220.
    1. Issa J. P. CpG island methylator phenotype in cancer. Nature Reviews Cancer. 2004;4(12):988–993. doi: 10.1038/nrc1507.
    1. Kleer C. G., Cao Q., Varambally S., et al. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proceedings of the National Acadamy of Sciences of the United States of America. 2003;100(20):11606–11611. doi: 10.1073/pnas.1933744100.
    1. Varambally S., Dhanasekaran S. M., Zhou M., et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419(6907):624–629. doi: 10.1038/nature01075.
    1. Kelley D. Z., Flam E. L., Izumchenko E., et al. Integrated analysis of whole-genome ChIP-Seq and RNA-Seq data of primary head and neck tumor samples associates HPV integration sites with open chromatin marks. Cancer Research. 2017;77(23):6538–6550. doi: 10.1158/0008-5472.CAN-17-0833.
    1. Bhasin J., Lee B., Matkin L., et al. Methylome-wide Sequencing Detects DNA Hypermethylation Distinguishing Indolent from Aggressive Prostate Cancer. Cell Reports. 2015;13(10):2135–2146. doi: 10.1016/j.celrep.2015.10.078.
    1. Shukla S., Kavak E., Gregory M., et al. CTCF-promoted RNA polymerase II pausing links DNA methylation to splicing. Nature. 2011;479(7371):74–79. doi: 10.1038/nature10442.
    1. Moarii M., Boeva V., Vert J.-P., Reyal F. Changes in correlation between promoter methylation and gene expression in cancer. BMC Genomics. 2015;16(1, article no. 873) doi: 10.1186/s12864-015-1994-2.
    1. Eun B., Sampley M. L., Good A. L., Gebert C. M., Pfeifer K. Promoter cross-talk via a shared enhancer explains paternally biased expression of Nctc1 at the Igf2/H19/Nctc1 imprinted locus. Nucleic Acids Research. 2013;41(2):817–826. doi: 10.1093/nar/gks1182.
    1. Sohni A., Bartoccetti M., Khoueiry R., et al. Dynamic switching of active promoter and enhancer domains regulates Tet1 and Tet2 expression during cell state transitions between pluripotency and differentiation. Molecular and Cellular Biology. 2015;35(6):1026–1042. doi: 10.1128/MCB.01172-14.
    1. Schwanhüusser B., Busse D., Li N., et al. Global quantification of mammalian gene expression control. Nature. 2011;473(7347):337–342. doi: 10.1038/nature10098.
    1. Kristensen A. R., Gsponer J., Foster L. J. Protein synthesis rate is the predominant regulator of protein expression during differentiation. Molecular Systems Biology. 2013;9:p. 689.
    1. Li J. J., Biggin M. D. Gene expression. Statistics requantitates the central dogma. Science. 2015;347(6226):1066–1067. doi: 10.1126/science.aaa8332.
    1. Jovanovic M., Rooney M. S., Mertins P., et al. Immunogenetics. Dynamic profiling of the protein life cycle in response to pathogens. Science. 2015;347(6226) doi: 10.1126/science.1259038.1259038
    1. Chen T., Lee C., Liu H., et al. APOBEC3A is an oral cancer prognostic biomarker in Taiwanese carriers of an APOBEC deletion polymorphism. Nature Communications. 2017;8(1) doi: 10.1038/s41467-017-00493-9.
    1. Zhang H., Liu T., Zhang Z. Integrated proteogenomic characterization of human high-grade serous ovarian cancer. Cell. 2016;166(3):755–765. doi: 10.1016/j.cell.2016.05.069.
    1. Mertins P., et al. Proteogenomics connects somatic mutations to signalling in breast cancer. Nature. 2016;534(7605):55–62.
    1. Zhang B., Wang J., Wang X. Proteogenomic characterization of human colon and rectal cancer. Nature. 2014;513(7518):382–387. doi: 10.1038/nature13438.
    1. Li L., Wei Y., To C., et al. Integrated Omic analysis of lung cancer reveals metabolism proteome signatures with prognostic impact. Nature Communications. 2014;5, article no. 5469 doi: 10.1038/ncomms6469.
    1. Shen H., et al. Integrated molecular characterization of testicular germ cell tumors. Cell Reports. 2018;23(11):3392–3406.
    1. Terunuma A., et al. MYC-driven accumulation of 2-hydroxyglutarate is associated with breast cancer prognosis. The Journal of clinical investigation. 2014;124(1):398–412.
    1. Turcan S., Rohle D., Goenka A., et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature. 2012;483(7390):479–483. doi: 10.1038/nature10866.
    1. Figueroa M. E., et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18(6):553–567. doi: 10.1016/j.ccr.2010.11.015.
    1. Auslander N., et al. A joint analysis of transcriptomic and metabolomic data uncovers enhanced enzyme-metabolite coupling in breast cancer. Scientific Reports. 2016;629662
    1. Ren S., et al. Integration of metabolomics and transcriptomics reveals major metabolic pathways and potential biomarker involved in prostate cancer. Molecular and cellular proteomics : MCP. 2016;15(1):154–163.
    1. Yang K., Xia B., Wang W., et al. A comprehensive analysis of metabolomics and transcriptomics in cervical cancer. Scientific Reports. 2017;7(1) doi: 10.1038/srep43353.43353
    1. Cancer Genome Atlas Research Network. Comprehensive and integrated genomic characterization of adult soft tissue sarcomas. Cell. 2017;171(4):950–965.e928
    1. Liu Y., et al. Comparative molecular analysis of gastrointestinal adenocarcinomas. Cancer Cell. 2018;33(4):721–735.e728
    1. Ricketts C. J., et al. The cancer genome atlas comprehensive molecular characterization of renal cell carcinoma. Cell Reports. 2018;23(12):p. 3698.
    1. Cancer Genome Atlas Research N. The Molecular Taxonomy of Primary Prostate Cancer. Cell. 2015;163(4):1011–1025.
    1. Cancer Genome Atlas Research N. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature. 2014;507(7492):315–322. doi: 10.1038/nature12965.
    1. Cancer Genome Atlas Research N. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513(7517):202–209.
    1. Cancer Genome Atlas Research N. Integrated genomic characterization of oesophageal carcinoma. Nature. 2017;541(7636):169–175. doi: 10.1038/nature20805.
    1. Cancer Genome Atlas Research N. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. The New England journal of medicine. 2013;368(22):2059–2074. doi: 10.1056/NEJMoa1301689.
    1. Cancer Genome Atlas N. Genomic Classification of Cutaneous Melanoma. Cell. 2015;161(7):1681–1696. doi: 10.1016/j.cell.2015.03.049.
    1. Cancer Genome Atlas Research N. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511(7511):543–550. doi: 10.1038/nature13879.
    1. Brennan C. W., Verhaak R. G., McKenna A., et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155:462–477. doi: 10.1016/j.cell.2013.09.034.
    1. Cerami E., Gao J., Dogrusoz U., et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discovery. 2012;2(5):401–404. doi: 10.1158/-12-0095.
    1. Werner H. M. J., Mills G. B., Ram P. T. Cancer systems biology: a peek into the future of patient care? Nature Reviews Clinical Oncology. 2014;11(3):167–176. doi: 10.1038/nrclinonc.2014.6.
    1. Guhathakurta D., Sheikh N. A., Meagher T. C., Letarte S., Trager J. B. Applications of systems biology in cancer immunotherapy: From target discovery to biomarkers of clinical outcome. Expert Review of Clinical Pharmacology. 2013;6(4):387–401. doi: 10.1586/17512433.2013.811814.

Source: PubMed

3
Tilaa