Whole genome sequencing of metastatic colorectal cancer reveals prior treatment effects and specific metastasis features

Pauline A J Mendelaar, Marcel Smid, Job van Riet, Lindsay Angus, Mariette Labots, Neeltje Steeghs, Mathijs P Hendriks, Geert A Cirkel, Johan M van Rooijen, Albert J Ten Tije, Martijn P Lolkema, Edwin Cuppen, Stefan Sleijfer, John W M Martens, Saskia M Wilting, Pauline A J Mendelaar, Marcel Smid, Job van Riet, Lindsay Angus, Mariette Labots, Neeltje Steeghs, Mathijs P Hendriks, Geert A Cirkel, Johan M van Rooijen, Albert J Ten Tije, Martijn P Lolkema, Edwin Cuppen, Stefan Sleijfer, John W M Martens, Saskia M Wilting

Abstract

In contrast to primary colorectal cancer (CRC) little is known about the genomic landscape of metastasized CRC. Here we present whole genome sequencing data of metastases of 429 CRC patients participating in the pan-cancer CPCT-02 study (NCT01855477). Unsupervised clustering using mutational signature patterns highlights three major patient groups characterized by signatures known from primary CRC, signatures associated with received prior treatments, and metastasis-specific signatures. Compared to primary CRC, we identify additional putative (non-coding) driver genes and increased frequencies in driver gene mutations. In addition, we identify specific genes preferentially affected by microsatellite instability. CRC-specific 1kb-10Mb deletions, enriched for common fragile sites, and LINC00672 mutations are associated with response to treatment in general, whereas FBXW7 mutations predict poor response specifically to EGFR-targeted treatment. In conclusion, the genomic landscape of mCRC shows defined changes compared to primary CRC, is affected by prior treatments and contains features with potential clinical relevance.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Size distributions of the different…
Fig. 1. Size distributions of the different types of structural variants.
Ridge-plot of the density of genomic sizes of structural variants in metastatic CRC. INV inversions (blue), DUP tandem duplications (purple), DEL deletions (orange). Source data are provided as a Source Data file.
Fig. 2. Oncoplot of metastatic CRC depicting…
Fig. 2. Oncoplot of metastatic CRC depicting identified driver genes and somatic mutations (SNV, InDels, and MNV).
Top panel: genes identified by dN/dS as driver genes per type of mutation; purple: frameshift variant; orange: other variant; blue: stop/gain variant; green: structural variant. Bottom panel: first track: clinical information: sex (male: orange; female: green) and second track: biopsy site. Track three (PLAT/PYR ± targeted) indicates which patients have been treated with platinum-based therapy (PLAT; e.g., oxaliplatin) and a pyrimidine-targeting drug (PYR; e.g., 5-FU), with or without the addition of another targeted treatment (±targeted; e.g., bevacizumab). Tracks four to six depict the distribution of the consensus molecular subtypes (CMS), tumor mutational burden (TMB), and the number of structural variant deletions of size 10kb–1Mb (DEL_CFS), partly associated with Common Fragile Sites (CFS), respectively. Source data are provided as a Source Data file.
Fig. 3. Mutational signatures in prior-treated cases…
Fig. 3. Mutational signatures in prior-treated cases compared to untreated cases.
Relative contribution (%) of several single and double base mutational signatures (SBS/DBS) in patients receiving prior treatment with platinum, pyrimidine antagonist, and targeted anti-EGFR treatment (PLAT/PYR + target; orange, n = 134) compared to untreated patients (blue, n = 124). Horizontal lines indicate the median. P-values are derived from the MWU test (two-sided) and corrected for multiple testing using the FDR (Hochberg) method. Source data are provided as a Source Data file.
Fig. 4. Mutational signatures in primary CRC…
Fig. 4. Mutational signatures in primary CRC and untreated metastatic CRC.
Relative contribution (%) of several single and double base mutational signatures (SBS/DBS) in primary CRC tumors (purple, n = 73), compared to untreated metastatic CRC tumors (green, n = 124). Horizontal lines indicate the median. P-values are derived from the MWU test (two-sided) and corrected for multiple testing using the FDR (Hochberg) method. Source data are provided as a Source Data file.
Fig. 5. Unsupervised hierarchical clustering of metastatic…
Fig. 5. Unsupervised hierarchical clustering of metastatic CRC using relative contribution of preselected mutational signatures.
Heatmap representing the median-centered relative contribution of mutational signatures between samples. Values were scaled from red (relative contribution above median) to yellow (relative contribution below median). Included single and doublet base signatures (SBS/DBS) are indicated at the right to which etiologies are added when known. Grouping of samples is shown by the dendrogram at the top. Source data are provided as a Source Data file.
Fig. 6. Actionable genes.
Fig. 6. Actionable genes.
Data from OncoKB were matched to affected genes observed in our mCRC cohort. Numbers indicate the number (and percentage) of affected patients. Source data are provided as a Source Data file.

References

    1. Atlas N. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487:330–337. doi: 10.1038/nature11252.
    1. Guinney J, et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 2015;21:1350–1356. doi: 10.1038/nm.3967.
    1. Lech G, Slotwinski R, Slodkowski M, Krasnodebski IW. Colorectal cancer tumour markers and biomarkers: recent therapeutic advances. World J. Gastroenterol. 2016;22:1745–1755. doi: 10.3748/wjg.v22.i5.1745.
    1. Ishaque N, et al. Whole genome sequencing puts forward hypotheses on metastasis evolution and therapy in colorectal cancer. Nat. Commun. 2018;9:4782. doi: 10.1038/s41467-018-07041-z.
    1. Haan JC, et al. Genomic landscape of metastatic colorectal cancer. Nat. Commun. 2014;5:5457. doi: 10.1038/ncomms6457.
    1. Yaeger R, et al. Clinical sequencing defines the genomic landscape of metastatic colorectal cancer. Cancer Cell. 2018;33(125–136):e123.
    1. Alexandrov LB, Nik-Zainal S, Wedge DC, Campbell PJ, Stratton MR. Deciphering signatures of mutational processes operative in human cancer. Cell Rep. 2013;3:246–259. doi: 10.1016/j.celrep.2012.12.008.
    1. Huyghe JR, et al. Discovery of common and rare genetic risk variants for colorectal cancer. Nat. Genet. 2019;51:76–87. doi: 10.1038/s41588-018-0286-6.
    1. Priestley P, et al. Pan-cancer whole-genome analyses of metastatic solid tumours. Nature. 2019;575:210–216. doi: 10.1038/s41586-019-1689-y.
    1. Kawakami H, Zaanan A, Sinicrope FA. Microsatellite instability testing and its role in the management of colorectal cancer. Curr. Treat. Options Oncol. 2015;16:30. doi: 10.1007/s11864-015-0348-2.
    1. Chen R, Sugiyama A, Seno H, Sugimoto M. Identification of modules and functional analysis in CRC subtypes by integrated bioinformatics analysis. PLoS ONE. 2019;14:e0221772. doi: 10.1371/journal.pone.0221772.
    1. Menghi F, et al. The tandem duplicator phenotype is a prevalent genome-wide cancer configuration driven by distinct gene mutations. Cancer Cell. 2018;34(197–210):e195.
    1. Li Y, et al. Patterns of somatic structural variation in human cancer genomes. Nature. 2020;578:112–121. doi: 10.1038/s41586-019-1913-9.
    1. Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature. 2010;465:1033–1038. doi: 10.1038/nature09144.
    1. Lanzós A, et al. Discovery of cancer driver long noncoding RNAs across 1112 tumour genomes: new candidates and distinguishing features. Sci. Rep. 2017;7:1–16. doi: 10.1038/srep41544.
    1. Li W, et al. Long non-coding RNA LINC00672 contributes to p53 protein-mediated gene suppression and promotes endometrial cancer chemosensitivity. J. Biol. Chem. 2017;292:5801–5813. doi: 10.1074/jbc.M116.758508.
    1. Alexandrov LB, et al. The repertoire of mutational signatures in human cancer. Nature. 2020;578:94–101. doi: 10.1038/s41586-020-1943-3.
    1. Blokzijl F, Janssen R, van Boxtel R, Cuppen E. MutationalPatterns: comprehensive genome-wide analysis of mutational processes. Genome Med. 2018;10:33. doi: 10.1186/s13073-018-0539-0.
    1. Christensen S, et al. 5-Fluorouracil treatment induces characteristic T>G mutations in human cancer. Nat. Commun. 2019;10:4571. doi: 10.1038/s41467-019-12594-8.
    1. Angus L, et al. The genomic landscape of metastatic breast cancer highlights changes in mutation and signature frequencies. Nat. Genet. 2019;51:1450–1458. doi: 10.1038/s41588-019-0507-7.
    1. Pleguezuelos-Manzano C, et al. Mutational signature in colorectal cancer caused by genotoxic pks(+) E. coli. Nature. 2020;580:269–273. doi: 10.1038/s41586-020-2080-8.
    1. Das D, Preet R, Mohapatra P, Satapathy SR, Kundu CN. 1, 3-Bis (2-chloroethyl)-1-nitrosourea enhances the inhibitory effect of resveratrol on 5-fluorouracil sensitive/resistant colon cancer cells. World J. Gastroenterol. 2013;19:7374. doi: 10.3748/wjg.v19.i42.7374.
    1. Zhang J, Sun W, Ren C, Kong X, Yan W, Chen X. A PolH transcript with a short 3′ UTR enhances PolH expression and mediates cisplatin resistance. Cancer Res. 2019;79:3714–3724. doi: 10.1158/0008-5472.CAN-18-3928.
    1. Tomicic MT, et al. Translesion polymerase η is upregulated by cancer therapeutics and confers anticancer drug resistance. Cancer Res. 2014;74:5585–5596. doi: 10.1158/0008-5472.CAN-14-0953.
    1. Kim J, et al. Somatic ERCC2 mutations are associated with a distinct genomic signature in urothelial tumors. Nat. Genet. 2016;48:600–606. doi: 10.1038/ng.3557.
    1. Strona G, Nappo D, Boccacci F, Fattorini S, San-Miguel-Ayanz J. A fast and unbiased procedure to randomize ecological binary matrices with fixed row and column totals. Nat. Commun. 2014;5:4114. doi: 10.1038/ncomms5114.
    1. Kondelin J, et al. Comprehensive evaluation of protein coding mononucleotide microsatellites in microsatellite-unstable colorectal cancer. Cancer Res. 2017;77:4078–4088. doi: 10.1158/0008-5472.CAN-17-0682.
    1. Boulagnon-Rombi C, et al. LRP1 expression in colon cancer predicts clinical outcome. Oncotarget. 2018;9:8849. doi: 10.18632/oncotarget.24225.
    1. Lupini L, et al. Prediction of response to anti-EGFR antibody-based therapies by multigene sequencing in colorectal cancer patients. BMC Cancer. 2015;15:808. doi: 10.1186/s12885-015-1752-5.
    1. Rachiglio AM, et al. Genomic profiling of KRAS/NRAS/BRAF/PIK3CA wild-type metastatic colorectal cancer patients reveals novel mutations in genes potentially associated with resistance to anti-EGFR agents. Cancers. 2019;11:859. doi: 10.3390/cancers11060859.
    1. Ready N, et al. First-line nivolumab plus ipilimumab in advanced non-small-cell lung cancer (CheckMate 568): outcomes by programmed death ligand 1 and tumor mutational burden as biomarkers. J. Clin. Oncol. 2019;37:992–1000. doi: 10.1200/JCO.18.01042.
    1. Massink MP, Kooi IE, Martens JW, Waisfisz Q, Meijers-Heijboer H. Genomic profiling of CHEK2*1100delC-mutated breast carcinomas. BMC Cancer. 2015;15:877. doi: 10.1186/s12885-015-1880-y.
    1. Canon J, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019;575:217–223. doi: 10.1038/s41586-019-1694-1.
    1. Barnes RP, Tsao WC, Moldovan GL, Eckert KA. DNA polymerase eta prevents tumor cell-cycle arrest and cell death during recovery from replication stress. Cancer Res. 2018;78:6549–6560. doi: 10.1158/0008-5472.CAN-17-3931.
    1. Li S, Wu X. Common fragile sites: protection and repair. Cell Biosci. 2020;10:1–9. doi: 10.1186/s13578-019-0370-3.
    1. Mermel CH, Schumacher SE, Hill B, Meyerson ML, Beroukhim R, Getz G. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 2011;12:R41. doi: 10.1186/gb-2011-12-4-r41.
    1. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics29, 15–21 (2013).
    1. Tarasov A, Vilella AJ, Cuppen E, Nijman IJ, Prins P. Sambamba: fast processing of NGS alignment formats. Bioinformatics. 2015;31:2032–2034. doi: 10.1093/bioinformatics/btv098.
    1. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30:923–930. doi: 10.1093/bioinformatics/btt656.
    1. Smid M, et al. Gene length corrected trimmed mean of M-values (GeTMM) processing of RNA-seq data performs similarly in intersample analyses while improving intrasample comparisons. BMC Bioinformatics. 2018;19:236. doi: 10.1186/s12859-018-2246-7.
    1. van Dessel LF, et al. The genomic landscape of metastatic castration-resistant prostate cancers reveals multiple distinct genotypes with potential clinical impact. Nat. Commun. 2019;10:1–13. doi: 10.1038/s41467-018-07882-8.
    1. Martincorena I, et al. Universal patterns of selection in cancer and somatic tissues. Cell. 2017;171(1029–1041):e1021.
    1. Canisius S, Martens JW, Wessels LF. A novel independence test for somatic alterations in cancer shows that biology drives mutual exclusivity but chance explains most co-occurrence. Genome Biol. 2016;17:261. doi: 10.1186/s13059-016-1114-x.
    1. Avvaru AK, Saxena S, Sowpati DT, Mishra RK. MSDB: a comprehensive database of simple sequence repeats. Genome Biol. Evol. 2017;9:1797–1802. doi: 10.1093/gbe/evx132.
    1. Eisenhauer EA, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Chakravarty, D. et al. OncoKB: a precision oncology knowledge base. JCO Precis. Oncol. (2017 Jul;2017:PO.17.00011. Epub 2017 May 16.).

Source: PubMed

3
Iratkozz fel