Whole genome sequencing analysis for cancer genomics and precision medicine

Hidewaki Nakagawa, Masashi Fujita, Hidewaki Nakagawa, Masashi Fujita

Abstract

Explosive advances in next-generation sequencer (NGS) and computational analyses have enabled exploration of somatic protein-altered mutations in most cancer types, with coding mutation data intensively accumulated. However, there is limited information on somatic mutations in non-coding regions, including introns, regulatory elements and non-coding RNA. Structural variants and pathogen in cancer genomes remain widely unexplored. Whole genome sequencing (WGS) approaches can be used to comprehensively explore all types of genomic alterations in cancer and help us to better understand the whole landscape of driver mutations and mutational signatures in cancer genomes and elucidate the functional or clinical implications of these unexplored genomic regions and mutational signatures. This review describes recently developed technical approaches for cancer WGS and the future direction of cancer WGS, and discusses its utility and limitations as an analysis platform and for mutation interpretation for cancer genomics and cancer precision medicine. Taking into account the diversity of cancer genomes and phenotypes, interpretation of abundant mutation information from WGS, especially non-coding and structure variants, requires the analysis of large-scale WGS data integrated with RNA-Seq, epigenomics, immuno-genomic and clinic-pathological information.

Keywords: cancer genome; mutational signature; non-coding mutation; structural variant; whole genome sequencing.

© 2018 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.

Figures

Figure 1
Figure 1
A, Whole genome sequencing (WGS) by next‐generation sequencer (NGS) can detect non‐coding mutations, structural variants (SV), including copy number alterations (CNA), mitochondria mutations and pathogen detection, as well as protein‐coding mutations; B, A representative Circos plot of cancer genome structure from WGS analysis, which indicates SV and CNA in all human chromosomes (1‐22+XY). Chromothripsis was observed in chromosomes 1 and 14. SNV, single nucleotide variants
Figure 2
Figure 2
A representative set of computational tools for cancer whole genome sequencing (WGS) analysis. As an initial step, raw sequence data (90‐150‐Gb ×2: FASTQ files) from next‐generation sequencer (NGS) of cancer genome and normal genome are aligned to the 3‐Gb human reference sequence (3 Gb), producing BAM files. PCR duplication is removed from the BAM file (usually a few percent). Somatic mutations are called by several types of algorithms specific to mutation types (SNV, short indels, CNA, SV and others), comparing variant allele numbers in cancer genomes with those in normal genomes by statistical analysis and creating a list of somatic mutations (VCF files). Germline variant call including SNV and indels is commonly performed from sequencing data of normal genomes using other software, HaplotypeCaller of GATK. SNV, single nucleotide variants; SV, structural variants
Figure 3
Figure 3
Non‐coding mutations and gene expression in whole genome sequencing (WGS) and RNA‐Seq. Intronic mutations can affect splicing forms. Mutations in 5′UTR and promoter regions can alter transcriptional activity, and regulatory elements such as enhancers, silencers or insulators in intergenic regions can affect chromatin structure and transcriptional activity. Mutations in 3′UTR can alter RNA stability and protein translation through changes in miRNA binding and other mechanisms. Mutations in non‐coding RNA, especially miRNA and lincRNA, may change the interaction of coding RNA/proteins and regulatory elements, and alter chromatin structure
Figure 4
Figure 4
Mutational signature and etiological factors in COSMIC database. The profile of each signature is displayed using the 6 substitution subtypes: C>A, C>G, C>T, T>A, T>C and T>G. Furthermore, each of the substitutions is examined by incorporating information on the bases immediately 5′ and 3′ to each mutated base, generating 96 possible mutation types. NMF analysis of cancer WGS in the COSMIC database (http://cancer.sanger.ac.uk/cosmic/signatures) demonstrates 30 mutational signatures at present, and 6 representative signatures are shown with their associated etiological factors for cancer development (aging, environmental exposures and defect of intrinsic DNA repair)

References

    1. Stratton M, Campbell PJ, Futreal A. The cancer genome. Nature. 2009;458:719‐724.
    1. Garraway LA, Lander ES. Lessons from the cancer genome. Cell. 2013;153:17‐37.
    1. Kinzler KW, Vogetstein B. Lessons from hereditary colorectal cancer. Cell. 1996;87:159‐170.
    1. King CR, Kraus M, Aaronson SA. Amplification of a novel v‐erbB related gene in human mammary carcinoma. Science. 1985;229:974‐976.
    1. Meyerson M, Gabriel S, Getz G. Advances in understanding cancer genomes through second‐generation sequencing. Nat Rev Genet. 2010;11:685‐696.
    1. Nakagawa H, Wardell CP, Furuta M, Taniguchi H, Fujimoto A. Cancer whole genome sequencing: present and future. Oncogene. 2015;34:5943‐5950.
    1. Cancer Genome Atlas Research Network . Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061‐1068.
    1. International Cancer Genome Consortium , Hudson TJ, Anderson W, et al. International network of cancer genome projects. Nature. 2010;464:993‐998.
    1. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome landscapes. Science. 2013;339:1546‐1558.
    1. Lowrence M, Stojanov P, Mermel C, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014;505:495‐501.
    1. Leiserson MD, Vandin F, Wu H, et al. Pan‐cancer network analysis identifies combinations of rare somatic mutations across pathways and protein complexes. Nat Genet. 2015;47:106‐114.
    1. Alexandrov LB, Nik‐Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415‐421.
    1. Laurence MS, Stojanov P, Polak P, et al. Mutational heterogeneity in cancer and the search for new cancer‐associated genes. Nature. 2013;499:214‐218.
    1. Forbes SA, Beare D, Gunasekaran P, et al. COSMIC: exploring the world's knowledge of somatic mutations in human cancer. Nucleic Acids Res. 2015;43:D805‐D811.
    1. Gnirke A, Melnikov A, Maguire J, et al. Solution hybrid selection with ultra‐long oligonucleotides for massively parallel targeted sequencing. Nat Biotechnol. 2009;27:182‐189.
    1. Bentley DR, Balasubramanian S, Swerdlow HP, et al. Accurate whole human genome sequencing using reversible terminator chemistry. Nature. 2008;456:53‐59.
    1. Treangen TJ, Salzberg SL. Repetitive DNA and next‐generation sequencing: computational challenges and solutions. Nat Rev Genet. 2011;13:36‐46.
    1. Eid J, Fehr A, Gray J, et al. Real‐time DNA sequencing form single polymerase molecule. Science. 2009;323:133‐138.
    1. Giordano F, Aigrain L, Quail MA, et al. De novo yeast genome assemblies from MinION, PacBio and MiSeq platforms. Sci Rep. 2017;7:3935.
    1. Dove ES, Joly Y, Tassé AM, et al. Genomic cloud computing: legal and ethical points to consider. Eur J Hum Genet. 2015;23:1271‐1278.
    1. Wang Q, Jia P, Li F, et al. Detecting somatic point mutations in cancer genome sequencing data: a comparison of mutation callers. Genome Med. 2013;5:91.
    1. Boutros PC, Ewing AD, Ellrott K, et al. Global optimization of somatic variant identification in cancer genomes with a global community challenge. Nat Genet. 2014;46:318‐319.
    1. Alioto TS, Buchhalter I, Derdak S, et al. A comprehensive assessment of somatic mutation detection in cancer using whole‐genome sequencing. Nat Commun. 2015;6:10001.
    1. Chen J, Weiss WA. Alternative splicing in cancer: implications for biology and therapy. Oncogene. 2015;34:1‐14.
    1. Xiong HY, Alipanahi B, Lee LJ, et al. The human splicing code reveals new insights into the genetic determinants of disease. Science. 2015;347:1254806.
    1. Shiraishi Y, Fujimoto A, Furuta M, et al. Integrated analysis of whole genome and transcriptome sequencing reveals diverse transcriptomic aberrations driven by somatic genomic changes in liver cancers. PLoS ONE. 2014;9:e114263.
    1. Supek F, Miñana B, Valcárcel J, Gabaldón T, Lehner B. Synonymous mutations frequently act as driver mutations in human cancers. Cell. 2014;156:1324‐1335.
    1. Zhao W, Pollack JL, Blagev DP, Zaitlen N, McManus MT, Erle DJ. Massively parallel functional annotation of 3′ untranslated regions. Nat Biotechnol. 2014;32:387‐391.
    1. Oikonomou P, Goodarzi H, Tavazoie S. Systematic identification of regulatory elements in conserved 3′ UTRs of human transcripts. Cell Rep. 2014;7:281‐292.
    1. FANTOM Consortium . A promoter‐level mammalian expression atlas. Nature. 2014;507:462‐470.
    1. Prensner JR, Chinnaiyan AM. The emergence of lncRNAs in cancer biology. Cancer Discov. 2011;1:391‐407.
    1. Fujimoto A, Furuta M, Totoki Y, et al. Whole genome mutational landscape and characterization of non‐coding and structural mutations in liver cancer. Nat Genet. 2016;48:500‐509.
    1. Rheinbay E, Parasuraman P, Grimsby J. Recurrent and functional regulatory mutations in breast cancer. Nature. 2017;547:55‐60.
    1. Freedman ML, Monteiro AN, Gayther SA, et al. Principles for the post‐GWAS functional characterization of cancer risk loci. Nat Genet. 2011;43:513‐518.
    1. ENCODE Project Consortium . An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57‐74.
    1. Roadmap Epigenomics Consortium . Integrative analysis of 111 reference human epigenomes. Nature. 2015;518:317‐330.
    1. Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA. Highly recurrent TERT promoter mutations in human melanoma. Science. 2013;339:957‐959.
    1. Vinagre J, Almeida A, Pópulo H, et al. Frequency of TERT promoter mutations in human cancers. Nat Commun. 2013;4:2185.
    1. Mansour MR, Abraham BJ, Anders L, et al. An oncogenic super‐enhancer formed through somatic mutation of a noncoding intergenic element. Science. 2014;346:1373‐1377.
    1. Fredriksson NJ, Ny L, Nilsson JA, Larsson E. Systematic analysis of noncoding somatic mutations and gene expression alterations across 14 tumor types. Nat Genet. 2014;46:1258‐1263.
    1. Weinhold N, Jacobsen A, Schultz N, Sander C, Lee W. Genome‐wide analysis of noncoding regulatory mutations in cancer. Nat Genet. 2014;46:1160‐1165.
    1. Katainen R, Kashyap DK, Pitkänen E, et al. CTCF/cohesion‐binding sites are frequently mutated in cancer. Nat Genet. 2015;47:818‐821.
    1. Canela A, Maman Y, Jung S, et al. Genome organization drives chromosome fragility. Cell. 2017;170:507‐521.
    1. Beroukhim R, Mermel CH, Porter D, et al. The landscape of somatic copy‐number alteration across human cancers. Nature. 2010;463:899‐905.
    1. Zack TI, Scumacher SE, Csarter SL, et al. Pan‐cancer patterns of somatic copy number alteration. Nat Genet. 2013;45:1134‐1140.
    1. Zhang X, Choi PS, Francis JM, et al. Identification of focally amplified lineage‐specific super‐enhancers in human epithelial cancers. Nat Genet. 2016;48:176‐182.
    1. Heitzer E, Ulz P, Belic J, et al. Tumor‐associated copy number changes in the circulation of patients with prostate cancer identified through whole‐genome sequencing. Genome Med. 2013;5:30.
    1. Wong FC, Lo YM. prenatal diagnosis innovation: genome sequencing of maternal plasma. Annu Rev Med. 2016;67:419‐432.
    1. Amant F, Verheecke M, Wlodarska I, et al. Presymptomatic identification of cancers in pregnant women during noninvasive prenatal testing. JAMA Oncol. 2015;1:814‐819.
    1. Oda Y, Tsuneyoshi M. Recent advances in the molecular pathology of soft tissue sarcoma: implications for diagnosis, patient prognosis, and molecular target therapy in the future. Cancer Sci. 2009;100:200‐208.
    1. Groffen J, Stephenson JR, Heisterkamp N, et al. Philadelphia chromosomal breakpoints are clustered within a limited region, bcr, on chromosome 22. Cell. 1984;36:93‐94.
    1. Soda M, Choi YL, Enomoto M, et al. Identification of the transforming EML4‐ALK fusion gene in non‐small‐cell lung cancer. Nature. 2007;448:561‐566.
    1. Kohno T, Ichikawa H, Totoki Y, et al. RET, ROS1 and ALK fusions in lung cancer. Nat Med. 2012;18:375‐377.
    1. Takeuchi K, Soda M, Togashi Y, et al. KIF5B‐RET fusions in lung adenocarcinoma. Nat Med. 2012;18:378‐381.
    1. Tomlins SA, Rhodes DR, Perner S, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005;310:644‐648.
    1. Northcott PA, Lee C, Zichner T, et al. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature. 2014;511:428‐434.
    1. Kataoka K, Shiraishi Y, Takeda Y, et al. Aberrant PD‐L1 expression through 3′‐UTR disruption in multiple cancers. Nature. 2016;534:402‐406.
    1. Cortes‐Ciriano I, Lee S, Park WY, Kim TM, Park PJ. A molecular portrait of microsatellite instability across multiple cancers. Nat Commun. 2017;8:15180.
    1. Lee E, Iskow R, Yang L, et al. Cancer Genome Atlas Research Network . Landscape of somatic retrotransposition in human cancers. Science. 2012;337:967‐971.
    1. Shukla R, Upton KR, Muñoz‐Lopez M, et al. Endogenous retrotransposition activates oncogenic pathways in hepatocellular carcinoma. Cell. 2013;153:101‐111.
    1. Kostic AD, Gevers D, Pedamallu CS, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012;22:292‐298.
    1. Geller LT, Barzily‐Rokni M, Danino T, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357:1156‐1160.
    1. Sung WK, Zheng H, Li S, et al. Genome‐wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat Genet. 2012;44:765‐769.
    1. Ojesina AI, Lichtenstein L, Freeman SS, et al. Landscape of genomic alterations in cervical carcinomas. Nature. 2014;506:371‐375.
    1. Cao S, Strong MJ, Wang X, et al. High‐throughput RNA sequencing‐based virome analysis of 50 lymphoma cell lines from the cancer cell line encyclopedia project. J Virol. 2015;89:713‐729.
    1. Cook LB, Melamed A, Niederer H, et al. The role of HTLV‐1 clonality, proviral structure and genomic integration site in adult T cell leukemia/lymphoma. Blood. 2014;123:3925‐3931.
    1. Zong WX, Rabinowitz JD, White E. Mitochondria and cancer. Mol Cell. 2016;61:667‐676.
    1. Reznik E, Miller ML, Şenbabaoğlu Y, et al. Mitochondrial DNA copy number variation across human cancers. Elife. 2016;5:pii: e10769.
    1. Stewart JB, Alaei‐Mahabadi B, Sabarinathan R, et al. Simultaneous DNA and RNA mapping of somatic mitochondrial mutations across diverse human cancers. PLoS Genet. 2015;11:e1005333.
    1. Ju YS, Tubio JM, Mifsud W, et al. Frequent somatic transfer of mitochondrial DNA into the nuclear genome of human cancer cells. Genome Res. 2015;25:814‐824.
    1. Bressac B, Kew M, Wands J, Ozturk M. Selective G to T mutations of p53 gene in hepatocellular carcinoma from southern Africa. Nature. 1991;350:29‐431.
    1. Alexandrov LB, Jones PH, Wedge DC, et al. Clock‐like mutational processes in human somatic cells. Nat Genet. 2015;47:1402‐1407.
    1. Poon SL, Pang T, McPherson JR, et al. Genome‐wide mutational signatures of aristolochic acid and its application as a screening tool. Sci Transl Med. 2013;5:197ra101.
    1. Alexandrov LB, Ju Y, Haase K, et al. Mutational signatures associated with tobacco smoking in human cancer. Science. 2016;354:618‐622.
    1. Nik‐Zainal S, Davies H, Staaf J, et al. Landscape of somatic mutations in 560 breast cancer whole‐genome sequences. Nature. 2016;534:47‐54.
    1. Korbel JO, Campbell PJ. Criteria for inference of chromothripsis in cancer genomes. Cell. 2013;152:1226‐1236.
    1. Rausch T, Jones DT, Zapatka M, et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell. 2012;148:59‐71.
    1. Waddell N, Pajic M, Patch A, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518:495‐501.
    1. Davies H, Glodzik D, Morganella S, et al. HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures. Nat Med. 2017;23:517‐525.
    1. Polak P, Karlić R, Koren A, et al. Cell‐of‐origin chromatin organization shapes the mutational landscape of cancer. Nature. 2015;518:360‐364.
    1. Ansell SM, Lesokhin AM, Borrello I, et al. PD‐1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N Engl J Med. 2015;372:311‐319.
    1. Davoli T, Uno H, Wooten EC, Elledge SJ. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science. 2017;355:eaaf8399.
    1. Shukla SA, Rooney MS, Rajasagi M, et al. Comprehensive analysis of cancer‐associated somatic mutations in class I HLA genes. Nat Biotechnol. 2015;33:1152‐1158.
    1. Zaretsky JM, Garcia‐Diaz A, Shin DS, et al. Mutations associated with acquired resistance to PD‐1 blockade in melanoma. N Engl J Med. 2016;375:819‐829.
    1. Dekker J, Marti‐Renom MA, Mirny LA. Exploring the three‐dimensional organization of genomes: interpreting chromatin interaction data. Nat Rev Genet. 2013;14:390‐403.

Source: PubMed

3
Abonnere