Relationship between genomic features and distributions of RS1 and RS3 rearrangements in breast cancer genomes.

Similar documents
Characterisation of structural variation in breast. cancer genomes using paired-end sequencing on. the Illumina Genome Analyser

A somatic mutational process recurrently duplicates germline susceptibility loci and tissue-specific super-enhancers in breast cancers

Supplementary Figure S1. Gene expression analysis of epidermal marker genes and TP63.

Nature Genetics: doi: /ng Supplementary Figure 1

Nature Genetics: doi: /ng Supplementary Figure 1. Mutational signatures in BCC compared to melanoma.

Broad H3K4me3 is associated with increased transcription elongation and enhancer activity at tumor suppressor genes

Nature Structural & Molecular Biology: doi: /nsmb.2419

Supplementary Figure 1: Features of IGLL5 Mutations in CLL: a) Representative IGV screenshot of first

SUPPLEMENTARY APPENDIX

Supplementary Information. Supplementary Figures

ChIP-seq data analysis

Supplementary Figures

Supplementary Figure 1: Attenuation of association signals after conditioning for the lead SNP. a) attenuation of association signal at the 9p22.

Plasma-Seq conducted with blood from male individuals without cancer.

Nature Neuroscience: doi: /nn Supplementary Figure 1

LTA Analysis of HapMap Genotype Data

Nature Genetics: doi: /ng Supplementary Figure 1. Somatic coding mutations identified by WES/WGS for 83 ATL cases.

Identification of Tissue Independent Cancer Driver Genes

Nature Genetics: doi: /ng Supplementary Figure 1. SEER data for male and female cancer incidence from

Whole Genome and Transcriptome Analysis of Anaplastic Meningioma. Patrick Tarpey Cancer Genome Project Wellcome Trust Sanger Institute

7SK ChIRP-seq is specifically RNA dependent and conserved between mice and humans.

Supplementary Figures

BWA alignment to reference transcriptome and genome. Convert transcriptome mappings back to genome space

Abstract. Optimization strategy of Copy Number Variant calling using Multiplicom solutions APPLICATION NOTE. Introduction

Expanded View Figures

Supplementary Figure 1. Efficiency of Mll4 deletion and its effect on T cell populations in the periphery. Nature Immunology: doi: /ni.

The Tandem Duplicator Phenotype Is a Prevalent Genome-Wide Cancer Configuration Driven by Distinct Gene Mutations

Unit 1 Exploring and Understanding Data

Nature Methods: doi: /nmeth.3115

Computational aspects of ChIP-seq. John Marioni Research Group Leader European Bioinformatics Institute European Molecular Biology Laboratory

Research Strategy: 1. Background and Significance

Nature Neuroscience: doi: /nn Supplementary Figure 1. Behavioral training.

Supplementary Figure 1. Copy Number Alterations TP53 Mutation Type. C-class TP53 WT. TP53 mut. Nature Genetics: doi: /ng.

Nature Medicine: doi: /nm.4439

Supplementary Materials for

Nature Getetics: doi: /ng.3471

ARTICLE RESEARCH. Macmillan Publishers Limited. All rights reserved

Journal: Nature Methods

Computer Science, Biology, and Biomedical Informatics (CoSBBI) Outline. Molecular Biology of Cancer AND. Goals/Expectations. David Boone 7/1/2015

Supplementary Figure 1

SUPPLEMENTARY INFORMATION

LESSON 3.2 WORKBOOK. How do normal cells become cancer cells? Workbook Lesson 3.2

Expanded View Figures

Nature Genetics: doi: /ng Supplementary Figure 1. HOX fusions enhance self-renewal capacity.

Analysis of Massively Parallel Sequencing Data Application of Illumina Sequencing to the Genetics of Human Cancers

Cancer Informatics Lecture

of TERT, MLL4, CCNE1, SENP5, and ROCK1 on tumor development were discussed.

Supplementary Figure 1: High-throughput profiling of survival after exposure to - radiation. (a) Cells were plated in at least 7 wells in a 384-well

Hands-On Ten The BRCA1 Gene and Protein

Supplemental Figure 1. Genes showing ectopic H3K9 dimethylation in this study are DNA hypermethylated in Lister et al. study.

Figure S2. Distribution of acgh probes on all ten chromosomes of the RIL M0022

Mapping by recurrence and modelling the mutation rate

Supplementary note: Comparison of deletion variants identified in this study and four earlier studies

Supplementary Figure 1: Comparison of acgh-based and expression-based CNA analysis of tumors from breast cancer GEMMs.

Supplementary results

Supplementary Figure 1. Estimation of tumour content

Genetic alterations of histone lysine methyltransferases and their significance in breast cancer

Chromatin marks identify critical cell-types for fine-mapping complex trait variants

Global variation in copy number in the human genome

Theta sequences are essential for internally generated hippocampal firing fields.

Introduction. Introduction

Expanded View Figures

Supplemental Figure legends

Sum of Neurally Distinct Stimulus- and Task-Related Components.

Genome-wide copy-number calling (CNAs not CNVs!) Dr Geoff Macintyre

RNA-Seq Preparation Comparision Summary: Lexogen, Standard, NEB

T. R. Golub, D. K. Slonim & Others 1999

Supplementary Tables. Supplementary Figures

Structural Variation and Medical Genomics

MIR retrotransposon sequences provide insulators to the human genome

High-order chromatin architecture determines the landscape of chromosomal alterations in cancer

Nature Immunology: doi: /ni Supplementary Figure 1. Characteristics of SEs in T reg and T conv cells.

Genomic structural variation

User Guide. Association analysis. Input

DOES THE BRCAX GENE EXIST? FUTURE OUTLOOK

Module 3: Pathway and Drug Development

Breast cancer. Risk factors you cannot change include: Treatment Plan Selection. Inferring Transcriptional Module from Breast Cancer Profile Data

Large-scale identity-by-descent mapping discovers rare haplotypes of large effect. Suyash Shringarpure 23andMe, Inc. ASHG 2017

Comparison of open chromatin regions between dentate granule cells and other tissues and neural cell types.

Single-strand DNA library preparation improves sequencing of formalin-fixed and paraffin-embedded (FFPE) cancer DNA

Detection of aneuploidy in a single cell using the Ion ReproSeq PGS View Kit

Supplementary Figure 1. Spectrum and signatures of substitutions.

Whole-genome detection of disease-associated deletions or excess homozygosity in a case control study of rheumatoid arthritis

University of Pittsburgh Annual Progress Report: 2008 Formula Grant

Supplementary information to:

Nature Biotechnology: doi: /nbt.1904

Lentiviral Delivery of Combinatorial mirna Expression Constructs Provides Efficient Target Gene Repression.

Frequency(%) KRAS G12 KRAS G13 KRAS A146 KRAS Q61 KRAS K117N PIK3CA H1047 PIK3CA E545 PIK3CA E542K PIK3CA Q546. EGFR exon19 NFS-indel EGFR L858R

White Paper Estimating Complex Phenotype Prevalence Using Predictive Models

OncoPPi Portal A Cancer Protein Interaction Network to Inform Therapeutic Strategies

Nature Genetics: doi: /ng Supplementary Figure 1

Supplementary Figure 1: Classification scheme for non-synonymous and nonsense germline MC1R variants. The common variants with previously established

Case Studies on High Throughput Gene Expression Data Kun Huang, PhD Raghu Machiraju, PhD

Interactive analysis and quality assessment of single-cell copy-number variations

bivariate analysis: The statistical analysis of the relationship between two variables.

Supplementary. properties of. network types. randomly sampled. subsets (75%

WDHS Curriculum Map Probability and Statistics. What is Statistics and how does it relate to you?

Numerous hypothesis tests were performed in this study. To reduce the false positive due to

Nature Genetics: doi: /ng Supplementary Figure 1. Assessment of sample purity and quality.

Transcription:

Supplementary Figure 1 Relationship between genomic features and distributions of RS1 and RS3 rearrangements in breast cancer genomes. (a,b) Values of coefficients associated with genomic features, separately for RS1 (a) and RS3 (b). The values of coefficients and 95% confidence intervals were obtained through negative binomial regression, where we divided the genome into 0.5-Mb bins. The panels show the exponentiated values, e wi, for ease of interpretation. The further a coefficient deviates from 1, the more it influences expected number of breakpoints in genomic regions.

Supplementary Figure 2 Tuning settings of the PCF algorithm for identification of hotspots. This summarizes the experiments conducted to gauge optimal parameters. Experiments were performed on observed data as well as simulations of rearrangements that took into account the background model of rearrangements. The x-axis indicates the setting of PCF parameters (g and i). The y-axis indicates the number of hotspots found in the observed (black dots) and simulated (grey dots) datasets. The blue rectangles highlight the PCF parameters that were finally selected to categorize hotspots of rearrangements in the observed data. The error bars at the grey dots denote standard deviation of the count when analysing 10 different simulated datasets. Red stars show estimated false discovery rate for the range of algorithm settings.

Supplementary Figure 3 Visualization of 33 hotspots of large (>100 kb) tandem duplications. The images display overlap of the rearrangements across the cohort, by showing cumulative number of samples with a tandem duplication involving each of the genomic regions. Dashed vertical lines represent boundaries of the hotspots. Thick red lines represent breast-tissue specific super enhancers. Blue vertical line represents position of germline susceptibility locus of breast cancer. Black lines above show positions of genes.

Supplementary Figure 4 Tandem-duplication hotspots are enriched in breast-tissue-specific super-enhancers and germline breast cancer susceptibility loci. (a) The likelihood of observing germline susceptibility loci coinciding with tandem duplication hotspots. Single-sided Poisson test. OR, odds ratio; error bars denote 95% confidence levels. (b) The likelihood of observing super-enhancers falling into tandem duplication hotspots. Density of breast-tissue specific super-enhancer and germline susceptibility loci for tandem duplication hotspots versus other tandemly duplicated regions that do not fall within hotspots. Single-sided Poisson test. OR, odds ratio; error bars denote 95% confidence levels. (c) Simulations were used to obtain an empirical null distribution of number of super-enhancer elements within the hotspots, presented as a histogram. We observed 59 super-enhancers in the hotspots. The likelihood of that observation according to the simulations is <0.0001.

Supplementary Figure 5 Enrichment of hotspots in breast-tissue super-enhancers and germline breast cancer susceptibility loci is robust with respect to the parameters of the PCF algorithm. The x-axis shows the parameter i of the PCF algorithm. First top panel shows which hotspot are detected at more stringent values of the i parameter. Second panel shows number of hotspots detected. Third and fourth panels depict the enrichments of breast cancer SNP loci and super-enhancers at more stringent values of the i parameter. Error bars denote 95% confidence intervals for the enrichment from Fisher s exact test.

Supplementary Figure 6 Relationship between tandem-duplicated segments and breast-tissue super-enhancer loci and germline breast cancer susceptibility SNP loci. In this analysis, all tandem duplication that had a breakpoint that fell within 1 Mb of super-enhancers (SENH, top panel) and/or breast cancer susceptibility SNPs (lower panel) were included. The x-axis reports on a 1-Mb genomic window surrounding SENH and SNPs, respectively. The y-axis reports the fraction of tandem duplications that have duplicated any given location within the 2-Mb window, out of all rearrangements in each group. The data are presented for RS1 tandem duplications in hotspots, RS1 tandem duplications that are not within hotspots and simulated RS1 rearrangements. Note the peak demonstrated for hotspot tandem duplication centered on the regulatory element/snp, which is not exhibited by tandem duplications that are not within hotspots or simulated data.

Supplementary Figure 7 Tandem duplications wholly or partially increase the number of copies of ESR1, which correlates with high expression of the gene. The top panel compares the expression of ESR1 between samples with and without tandem duplications in the hotspot. Samples that have tandem duplicated ESR1, even by just a single tandem duplication, have ESR1 expression levels that are in a similar high range as ER-positive tumors and are distinctly elevated when compared to the triple-negative tumors. The boxes highlight median expression level of the gene, with lower and upper quartiles. The second panel shows expression of ESR1 in individual samples with tandem duplications in the hotspot. The bottom panel shows the position of the rearrangements with respect to ESR1 gene body on the left, and across entire chromosome 6 on the right. Copy number (y-axis) depicted as black dots (10-kb bins). Green lines present tandem duplication breakpoints.

Supplementary Figure 8 Tandem duplications in some hotspots (wholly or partially) increase the number of copies of specific driver genes associated with breast cancer, even if by only one or two copies. Left shows focus on the hotspot. Right shows entire chromosome of the hotspot. Rows correspond to individual samples. Copy number (y-axis) depicted as black dots (10-kb bins). Green lines present tandem duplication breakpoints. The ZNF217 locus is an example of a tandem duplication hotspot. Each patient has an apparent increase in copy number through a long tandem duplication, wholly of the gene. This site is enriched for breast tissue-specific super-enhancers.

Supplementary Figure 9 Tandem duplications in the hotspots are a feature of samples with many or few rearrangements in their genomes. A histogram of the frequency of each of the 33 RS1-enriched tandem duplication hotspots is shown in the topmost panel with the 33 hotspots noted across the horizontal axis. The number of samples with rearrangements within any of the 33 hotspots is noted on the vertical axis on the left. A histogram of the number of hotspots per sample is provided on the right (purple, BRCA1-intact HR-deficient cancers; blue, BRCA1-null HR-deficient cancers; black, all other groups). Central matrix depicts the relationship between samples and number of hotspots (black, hotspot rearrangement present).

Supplementary Figure 10 Expression of MYC in samples with and without tandem duplications in the hotspot, distinguishing among breast cancer subtypes. The boxes highlight median expression level of the gene, with lower and upper quartiles. These data were used to fit a linear model, suggesting that a tandem duplication in the hotspot was correlated with increased expression of the gene by 0.99 log 2 FPKM, with P = 4.4 x 10-4.

Supplementary Figure 11 Hotspots of tandem duplications can be detected only in cohorts with an adequate number of rearrangements. We sub-sampled the rearrangement dataset from the breast cancer cohort, in order to assess how many hotspots we could have detected in smaller cohorts. The number of RS1 rearrangements in the ovarian cohort was sufficient to detect hotspots, and indeed, in the ovarian cohort we detected seven hotspots. The number of rearrangements in pancreatic cohort was insufficient to detect hotspots, and indeed we detected none there.

Supplementary Figure 12 A visualization of the RS1 hotspots in ovarian cancers. The images display overlap of the rearrangements across the cohort, by showing cumulative number of samples with a tandem duplication involving each of the genomic regions. Thick red lines represent ovarian-tissue specific super enhancers. Black lines above show positions of genes. Dashed vertical lines represent boundaries of the hotspots.

Supplementary Note 1. Modelling the background distribution of rearrangements Rearrangements are known to have an uneven distribution in the genome. There have been numerous descriptions linking genomic features such as replication timing with the non-uniform distribution of rearrangements. Thus, any analysis that seeks to detect regions of higher mutability than expected must take the genomic features that influence this non-uniform distribution into account in its background model. In order to formally detect and quantify associations between genomic features and somatic rearrangements in breast cancer, we conducted a multi-variate genome-wide regression analysis. The genome was divided into non-overlapping genomic bins of 0.5 Mb, and each bin was characterised for the following genomic features: replication time domain as determined using Repli-Seq data from the MCF7 breast cancer cell line (ENCODE) gene expression levels o highly expressed genes (top 25% of genes when ranked by average expression level in our cohort) o low-expressed genes (remaining 75% of genes) copy number: average total copy number across the bin in the cohort repetitive sequences: o Segmental duplications o ALU elements o Other types of repeats DNAse hyper-sensitive sites (peaks, MCF7, Encode) Non-mapping sites: N bases in the reference genome Known fragile sites (Bignell et al., 2010) Chromatin staining

All of the above features were normalised to a mean of 0 and standard deviation of 1 across the bins for each feature, in order to permit comparability between features. The total number of RS1 and RS3 rearrangement breakpoints were counted for each bin. A regression model was performed in order to learn associated features, using a negative binomial distribution to account for potential over-dispersion. The model was trained on a total 4,481 bins, after removing the bins containing validated cancer genes. We found that features such as early replication time, highly expressed genes, elevated (general) copy number, DNAse1 hypersensitivity sites and ALU elements were associated with higher densities of RS1 and RS3 rearrangements (Supplementary Figure S1). They were similarly associated for both tandem duplication signatures although absolute levels of enrichment were only slightly different between the two. Of note, features such as fragile sites, chromatin staining, many classes of repeat elements were neither significantly enriched nor de-enriched for RS1 or RS3 rearrangements. The properties learned through this regression analysis were then used to perform simulations of rearrangements as described in the next sections, and to calculate the expected number of breakpoints in regions of the genome depending on their features. Given genomic features of a bin f i (there are N such features) and weights of the negative binomial regression w i, and intercept m, the expected number of breakpoints in a bin given by: b i = e m N e w if i i=1 In Supplementary Figure S1 we show the exponentiated parameters e m and e w i fitted by the model, as in this form they have an intuitive multiplicative interpretation. If e w i = 1, the i th genomic feature does not affect the expected number of breakpoints in bins.

2. Simulations of rearrangements Simulations consisted of as many rearrangements as was observed for each sample in the dataset, preserving the type of rearrangement (tandem duplication, inversion, deletion or translocation), the length of each rearrangement (distance between partner breakpoints) and ensuring that both breakpoints fell within mappable/callable regions in our pipeline. Simulations also took into account the genomic bias of rearrangements that were identified above. In other words, for each rearrangement that was simulated, we: Drew a position for the lower breakpoint from a genomic bin. Sampling of the lower bin was weighted (non-uniform), with weights proportional to b i, the expected number of breakpoint in each bin according to the background model. Within that bin, we uniformly sampled a random genomic position. Drew the partner breakpoint at an equivalent length as was observed for that rearrangement The procedure was repeated 10,000 times to build a null distribution. Genomic biases of simulated rearrangements have been confirmed to behave in a similar way to the observed biases. This null distribution served as the comparator for the next set of analyses, where we used a segmentation algorithm to detect regions that are more mutable than would be expected from our simulations, which correct for the genomic properties that we know influence the uneven distribution of rearrangements. 3. Interpretation of the RS1 and RS3 hotspots at the NEAT1/MALAT1 locus

Notably, the RS3 hotspot at NEAT1/MALAT1 is the only hotspot that is also an RS1 hotspot. 17 samples contributed to the RS3 hotspot at the site, yet no pattern of effect was noted. Neither MALAT1 nor NEAT1 were transected by the RS3 rearrangements. On the contrary, a clearer pattern was apparent among the samples with RS1 rearrangements. Out of the eight samples that had RS1 rearrangements in the hotspot, we observed a duplication of either NEAT1 or MALAT1 in seven samples. In all eight samples the RS1 duplication spanned one of the three super-enhancers nearby. Intriguingly, these lncrnas were also identified as being hotspots for indel and substitution mutagenesis in an experiment searching for putative non-coding drivers (Nik-Zainal, 2016b). We find that the distribution of indel sizes in this region is out-of-keeping with the general distribution of indels in breast cancers. Most were microhomology-mediated indels, which would have commenced as double-strand breaks (DSB) and been fixed latterly by microhomology-mediated end joining mechanisms. NEAT1 and MALAT1 are two of the most highly expressed lncrnas in breast tissue. Thus, the observation that this is a hotspot of different rearrangement signatures and an indel signature, all of which would have started as DSBs that were eventually fixed using different compensatory DSB repair pathways, would suggest that this is simply a site that is highly exposed to damage. This is likely to be because it is one of the more highly transcribed sites in breast tissue. This interpretation would suggest that the clustering of mutations observed here is not due to selective pressure and that these mutations are not driver events. However, this does not preclude highly significant physiological roles for NEAT1/MALAT1 in the development of cancer. Indeed, it would appear that it is because of the very important biological roles played by NEAT1/MALAT1 that they could be extremely highly transcribed and thus selectively susceptible to DSB mutagenesis.

4. Identifying hotspots for remaining rearrangement signatures, other than RS1 and RS3 Of the six rearrangement signatures, RS4 and RS6 are characterised by interchromosomal and intrachromosomal clustered rearrangements respectively, and RS2 is defined by dispersed interchromosomal rearrangements. RS5 consists mostly of dispersed deletions, mainly shorter than 10 kb. We hypothesised that distribution of the other rearrangements signatures, particularly the clustered rearrangements, is strongly affected by selection, and we did not build their background models. For these signatures, their genomewide rearrangement densities served as expected densities in each segment. As hotspots of these signatures the PCF algorithm identified regions with breakpoint density higher than the neighbouring regions and at least twice the genome-wide density. The hotspots of signatures RS2, RS4, RS5, and RS6 are listed and annotated in Supplementary Table S3. RS4 and RS6 signatures demonstrated 13 hotspots each, 8 of which were overlapping with each other and coincided with various well-described driver amplicons including ERBB2, IGF1R, CCND1, chr8:znf703/fgfr1 and ZNF217. Similarly, RS2 demonstrated 21 loci, many of which fell within driver amplicon loci or coincided with known retrotransposition loci. RS5 is characterised by deletion rearrangements and only 3 hotspots were identified, all of which likely represented putative driver loci (PTEN, QKI and TRPS1). 5. Analysis of gene expression RNA expression levels of genes in the samples were obtained from RNA-seq data as reported by another publication (Nik-Zainal, 2016a). We set out to assess whether tandem duplications in the hotspots are associated with increased

expression of affected genes. Statistical methods and results are presented in Supplementary Note Section 3. We set out to assess whether tandem duplications in the hotspots are associated with increased expression of affected genes. However, in many instances, the number of samples contributing to a specific hotspot that also had transcriptomic data was a limiting factor. For example, only six out of fourteen samples that contributed to the ESR1 hotspot had transcriptomic data available. c-myc however was a commonly affected locus that had an adequate number of samples (12 samples in the hotspot of which 4 had tandem duplications of the gene itself ) to use a linear model to assess the correlation between presence of RS1 tandem duplications at the loci, and the gene expression level, while accounting for different breast receptor expression subtypes (ER positive, triple negative, HER2 positive) and their baseline copy number (background copy number can be variable from one part of the genome to the next e.g. whole arm gains or losses across the genome, or large amplicons). The model was given by: e ~ r + c + t where e : gene expression log2 FPKM r : receptor type of a sample: ER positive, triple negative, HER2 positive c log2 of background copy number of the gene in individual samples; if the gene itself was tandem duplicated by a dispersed rearrangement, we count the copy number outside of the duplication t : whether tandem duplications are present in nearby hotspot: TRUE/FALSE The regression model accounts for the variation in gene expression due to amplifications through the parameter c. To establish the effect of tandem duplications on gene expression, we estimate the value of coefficient t.

We obtained the estimates of coefficients in the regression model. We find that the tandem duplications at the c-myc hotspot are significantly associated with the expression of MYC. On average, a tandem duplication within the hotspot corresponds to an increase of the gene by 0.99 log2 FPKM (P=4.4 10-4 in t-test, t-value 3.56). In other words, tandem duplications within a c-myc hotspot were associated with an increase in c-myc expression level of 2 FPKM (Supplementary Table 5). The ability to explore expression effects of tandem duplications of superenhancers or breast cancer susceptibility SNP loci was limited by the fact that downstream targets of these putative regulatory elements are frequently unknown, uncertain and/or usually involving multiple genes rather than simply a single downstream effector. We thus took a global gene expression approach, to permit detection of expression effects across many genes. This method has its limitations - true signal in some genes may be diluted by the noise from many other genes that are not contributing any signal. However, it does permit detection of effects from many genes simultaneously. In order to account for between gene variation and tumour subtypes, we used the following mixed-effects linear model: e ~ (1 gene) + (r gene) + c + dg + ds + do where: e : gene expression log2 FPKM random components: (1 gene) : intercept which is different for each gene (r gene) : asjustment for receptor type of a sample (ER+, TN, HER2+) which may be different between genes fixed components: c copy number of the gene in a sample from ASCAT (log2) dg : whether the gene was tandem duplicated

ds: whether a super-enhancer or a breast cancer susceptibility locus within 1Mb of the gene was tandem duplicated (the categories are mutually exclusive, so if a duplication covers both a gene and the super-enhancer, it will appear in the former category only) do : whether there is some other tandem duplication within 1Mb In order to assess the statistical significance of the associations, we also defined two null models. The first one allows us to see and quantify the effects of the tandem duplications of breast cancer super-enhancer or breast cancer susceptibility SNP loci. The first one allows us to see and quantify the effects of tandem duplications of genes themselves. Null model 1: Null model 2: e ~ (1 gene) + (r gene) + c + dg + do e ~ (1 gene) + (r gene) + c + ds + do P-values were obtained by likelihoods ratio tests, between the full and null models, using ANOVA. For fitting the models, we used R and lme4. We were able to assess the association between tandem duplications in the hotspots and expression levels of different groups of genes including: 13 putative oncogenes that are implicated in these hotspots: ETV6, MDM2, SRGAP3, WWTR1, FGFR3, WHSC1, MYC, NOTCH1, ESR1, FOXA1, MAML2, ERBB2, ZNF217. Remaining 509 genes in the hotspots. A random selection of 489 genes outside of the hotspots We report all of the coefficients of the regression models in Supplementary Table 5. In general, tandem duplications in the hotspots were associated with increases in expression levels of nearby genes.

A tandem duplication of an oncogene would be associated with an average increase of expression levels by 0.58 log2 FPKM (standard error 0.17, P=6.3 10-4, by likelihood-ratio test with null model 2, chisq=11.697, 12 and 13 degrees of freedom in the two compared models). A tandem duplication of a super-enhancer or regions containing a breast cancer susceptibility SNP proximal to the gene, but not the gene itself, would be associated with an average increase of expression levels of oncogenes by 0.30 (s.e. 0.20) (P=0.12, by likelihood-ratio test with null model 1, chisq=2.3491, 12 and 13 degrees of freedom in the two compared models) A tandem duplication of any of the remaining 509 genes in the RS1 hotspots (not the oncogenes listed) would be associated with their average increase of expression levels by 0.45 log2 FPKM (s.e. 0.03) (P=2.2 10-16, in likelihood-ratio test with null model 2, chisq=195.7, 13 and 14 degrees of freedom in the two compared models). A tandem duplication of a super-enhancer or regions containing a breast cancer susceptibility SNP proximal to the gene, but not the gene itself, would be associated with an average increase of expression levels of the 509 genes by 0.16 (s.e. 0.04) (P=1.8 10-4 by likelihood-ratio test with null model 1, chisq=14.037, 13 and 14 degrees of freedom in the two compared models). 6. Hotpots of RS1 in other tumours In addition to breast cancer, tumours of other tissue types sometimes show excess of tandem duplications in their genomes. In order to investigate whether the rearrangements in other tumor types also accumulate in hotspots, we utilized previously published sequences of ovarian and pancreatic cancer genomes. We investigated whether the hotspots would also co-localize with tissue specific super-enhancers.

We analyzed data from 73 ovarian and 96 pancreatic cancers. Applying the same algorithms as for the breast cancer, we identified 2,923 RS1 rearrangements in ovarian cohort and 448 in pancreatic (compared to 5,944 in breast cancer cohort). In order to assess how many rearrangements are needed to detect hotspots, we randomly sub-sampled the rearrangement dataset from breast cancer, and we present results from the simulation in Supplementary Figure S11. The results from the simulation matched the number of hotspots detected in ovarian and pancreatic data. We did not find any hotspots in the pancreatic cancer data, and we would have detected none in the breast cancer dataset either, with the same number of tandem duplications as shown in the simulations. However, we were able to identify 7 hotspots of RS1 rearrangements in the ovarian cancer cohort, also consistent with the simulations. We fitted a background model to the ovarian rearrangements using the copy number data specific to ovarian samples, and applied the PCF algorithm with identical parameters. We identified 7 hotspots of RS1 signature, only one of which coincided with the hotspots we had identified in the breast tumours (RS1_OV_chr3_48.6Mb). Please refer to Supplementary Table S6 for the coordinates of the RS1 hotspots in ovarian cancers, and Supplementary Figure S12 for their visualization. The enrichment of ovarian super-enhancers in the hotspots compared to rest of tandem-duplicated genome was 2.90 fold. MUC1 was focally tandem duplicated in one of the ovarian hotspots (RS1_OV_chr1_150.3Mb). 7. Supplementary tables Table S1: Hotspots of rearrangement signatures RS1 and RS3 identified through PCF-based method. A, Description of headers. B, Summary of hotspots.

Table S2. Genomic consequences of RS1 and RS3 duplications, related to Main Figure 4. Numbers of duplications and transections of genomic elements, separately for RS1 and RS3, inside and outside of the hotspots. Table S3: Hotspots of other rearrangement signatures (RS2, RS4, RS5, RS6) identified through PCF-based method. A, Description of headers. B, Summary of hotspots. Table S4. Genomic features of the RS1 hotspots. Comparison with the rest of tandem-duplicated genome with respect to: breast cancer susceptibility SNPs, breast tissue super-enhancers, non-breast super-enhancers, known oncogenes, promoters, enhancers, broad fragile sites, narrow fragile sites. A, Description of headers. B, Associations. Table S5: Modelling the effects of RS1 tandem duplications on gene expression. Rows coefficients used in the regression models. Columns experiments with different sets of genes. In the table we show the fitted values of regression coefficients. Table S6: Hotspots of rearrangement signatures RS1 and RS3 identified through PCF-based method in ovarian tumours. A, Description of headers. B, Summary of hotspots.