The Sweden Cancerome Analysis Network - Breast (SCAN-B) Initiative: a large-scale multicenter infrastructure towards implementation of breast cancer genomic analyses in the clinical routine

Lao H Saal, Johan Vallon-Christersson, Jari Häkkinen, Cecilia Hegardt, Dorthe Grabau, Christof Winter, Christian Brueffer, Man-Hung Eric Tang, Christel Reuterswärd, Ralph Schulz, Anna Karlsson, Anna Ehinger, Janne Malina, Jonas Manjer, Martin Malmberg, Christer Larsson, Lisa Rydén, Niklas Loman, Åke Borg, Lao H Saal, Johan Vallon-Christersson, Jari Häkkinen, Cecilia Hegardt, Dorthe Grabau, Christof Winter, Christian Brueffer, Man-Hung Eric Tang, Christel Reuterswärd, Ralph Schulz, Anna Karlsson, Anna Ehinger, Janne Malina, Jonas Manjer, Martin Malmberg, Christer Larsson, Lisa Rydén, Niklas Loman, Åke Borg

Abstract

Background: Breast cancer exhibits significant molecular, pathological, and clinical heterogeneity. Current clinicopathological evaluation is imperfect for predicting outcome, which results in overtreatment for many patients, and for others, leads to death from recurrent disease. Therefore, additional criteria are needed to better personalize care and maximize treatment effectiveness and survival.

Methods: To address these challenges, the Sweden Cancerome Analysis Network - Breast (SCAN-B) consortium was initiated in 2010 as a multicenter prospective study with longsighted aims to analyze breast cancers with next-generation genomic technologies for translational research in a population-based manner and integrated with healthcare; decipher fundamental tumor biology from these analyses; utilize genomic data to develop and validate new clinically-actionable biomarker assays; and establish real-time clinical implementation of molecular diagnostic, prognostic, and predictive tests. In the first phase, we focus on molecular profiling by next-generation RNA-sequencing on the Illumina platform.

Results: In the first 3 years from 30 August 2010 through 31 August 2013, we have consented and enrolled 3,979 patients with primary breast cancer at the seven hospital sites in South Sweden, representing approximately 85% of eligible patients in the catchment area. Preoperative blood samples have been collected for 3,942 (99%) patients and primary tumor specimens collected for 2,929 (74%) patients. Herein we describe the study infrastructure and protocols and present initial proof of concept results from prospective RNA sequencing including tumor molecular subtyping and detection of driver gene mutations. Prospective patient enrollment is ongoing.

Conclusions: We demonstrate that large-scale population-based collection and RNA-sequencing analysis of breast cancer is feasible. The SCAN-B Initiative should significantly reduce the time to discovery, validation, and clinical implementation of novel molecular diagnostic and predictive tests. We welcome the participation of additional comprehensive cancer treatment centers.

Trial registration: ClinicalTrials.gov identifier NCT02306096.

Figures

Figure 1
Figure 1
Overview of the SCAN-B infrastructure. Shown are the SCAN-B clinical (green boxes), laboratory (blue), and computational and analytical (orange) components. Solid black arrows indicate flow of material, and dashed black lines indicate flow of information. Enrollment and sampling of patients at time of preoperative (neoadjuvant) biopsy is not shown. ds, double-stranded; INCA, Swedish national breast cancer registry; TMA, tissue microarray.
Figure 2
Figure 2
Study demographics and clinical variables. (A) For the period 30 August 2010 to 31 August 2013, yearly (non-calendar) summary of the number of enrolled patients, the number of patients with preoperative blood sample collected, and number of patients with tumor specimen collected. (B-H) For the two complete calendar years 2011 and 2012 that could be matched to the INCA Swedish national breast cancer registry, (B) chart of all cases with a preoperative diagnosis of primary breast cancer within the catchment region divided into those that were accrued or not accrued. Comparison of baseline clinical variables between all eligible breast cancer patients, patients accrued, and patients accrued with tumor sample, for (C) estrogen receptor (ER) status, (D) progesterone receptor (PgR) status, (E) HER2 status, (F) age at diagnosis, (G) Nottingham histological grade (NHG), and (H) tumor size. †,‡ Significant differences were identified between all diagnoses and accrued with tumor specimen for NHG (P = 0.005) and tumor size (P <0.001), and between patients accrued and accrued with biopsy for NHG (P = 0.025) and tumor size (P <0.001).
Figure 3
Figure 3
RNA sequencing and microarray analysis for population-based breast tumors. (A) Hierarchical clustering of 49 primary breast tumors (clustered columns) using the RNA-seq gene expression measurements and the PAM50 intrinsic gene signature (clustered rows). Clinical annotations for estrogen receptor (ER), progesterone receptor (PgR), and HER2 are indicated below the sample dendrogram, and PAM50 intrinsic subtyping is shown for classification using RNA-seq data as well as using microarray data generated from the same input RNA (90% concordant; results for Sørlie (92%) and Hu (96%) signatures are presented in Additional file 2: Figure S2). Genes of interest are highlighted in red, and relative expression level is indicated by the box color (see color key below the heatmap). For six tumor samples, technical replicates from the same RNA sources were performed for both RNA-seq and microarrays; plotted in (B) and (C) are representative examples comparing the fold-change for all RefSeq genes between two tumors (Y axis), and the fold-change between the replicated experiments for the same two tumors (X axis). Consistently, RNA-seq demonstrated values closer to the ideal line of identity and for a broader dynamic range. The +/- 2 fold-change (|log2| = 1) thresholds are indicated by blue dashed lines. (D) RNA-seq-derived expression level of ESR1, which encodes the ER alpha protein, is shown compared to the clinical ER IHC score for each of the 49 tumors. See Additional file 2: Figure S3 for corresponding plots for progesterone receptor and ERBB2 (HER2).
Figure 4
Figure 4
Detection of mutations by RNA-seq. (A) Eighteen genes with at least one mutation (out of 90 genes screened) across the 49 population primary breast tumors are shown, in order of frequency (see totals and percentages to the right of each gene row). Mutant allele frequency is indicated by the box color (see key below matrix). All mutations are non-synonymous missense mutations except those indicated by F (frameshift) and X (nonsense). Tumor sample dendrogram is as in Figure 3A. Predicted mutant amino acids are shown for (B)PIK3CA which encodes the p110-alpha catalytic subunit of the phosphatidylinositol-4,5-bisphosphate 3-kinase oncogene, and (C)TP53 which encodes the tumor suppressor TP53.

References

    1. Engholm G, Ferlay J, Christensen N, Bray F, Gjerstorff ML, Klint A, et al. NORDCAN–a Nordic tool for cancer information, planning, quality control and research. Acta Oncol. 2010;49:725–36. doi: 10.3109/02841861003782017.
    1. Coleman MP, Forman D, Bryant H, Butler J, Rachet B, Maringe C, et al. Cancer survival in Australia, Canada, Denmark, Norway, Sweden, and the UK, 1995-2007 (the International Cancer Benchmarking Partnership): an analysis of population-based cancer registry data. Lancet. 2011;377:127–38. doi: 10.1016/S0140-6736(10)62231-3.
    1. Brenner H, Hakulinen T. Very-long-term survival rates of patients with cancer. J Clin Oncol. 2002;20:4405–9. doi: 10.1200/JCO.2002.99.060.
    1. Dodwell D, Thorpe H, Coleman R. Refining systemic therapy for early breast cancer: difficulties with subtraction. Lancet Oncol. 2009;10:738–9. doi: 10.1016/S1470-2045(09)70203-7.
    1. Gordon L, Scuffham P, Hayes S, Newman B. Exploring the economic impact of breast cancers during the 18 months following diagnosis. Psychooncology. 2007;16:1130–9. doi: 10.1002/pon.1182.
    1. Armstrong K. Can genomics bend the cost curve? JAMA. 2012;307:1031–2. doi: 10.1001/jama.2012.261.
    1. Meyerson M, Gabriel S, Getz G. Advances in understanding cancer genomes through second-generation sequencing. Nat Rev Genet. 2010;11:685–96. doi: 10.1038/nrg2841.
    1. Van’t Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415:530–6. doi: 10.1038/415530a.
    1. Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004;351:2817–26. doi: 10.1056/NEJMoa041588.
    1. Ahmed AA, Brenton JD. Microarrays and breast cancer clinical studies: forgetting what we have not yet learnt. Breast Cancer Res. 2005;7:96–9. doi: 10.1186/bcr1017.
    1. Reis-Filho JS, Westbury C, Pierga JY. The impact of expression profiling on prognostic and predictive testing in breast cancer. J Clin Pathol. 2006;59:225–31. doi: 10.1136/jcp.2005.028324.
    1. Hedenfalk I, Duggan D, Chen Y, Radmacher M, Bittner M, Simon R, et al. Gene-expression profiles in hereditary breast cancer. N Engl J Med. 2001;344:539–48. doi: 10.1056/NEJM200102223440801.
    1. Gruvberger S, Ringner M, Chen Y, Panavally S, Saal LH, Borg A, et al. Estrogen receptor status in breast cancer is associated with remarkably distinct gene expression patterns. Cancer Res. 2001;61:5979–84.
    1. Saal LH, Johansson P, Holm K, Gruvberger-Saal SK, She QB, Maurer M, et al. Poor prognosis in carcinoma is associated with a gene expression signature of aberrant PTEN tumor suppressor pathway activity. Proc Natl Acad Sci U S A. 2007;104:7564–9. doi: 10.1073/pnas.0702507104.
    1. Staaf J, Ringner M, Vallon-Christersson J, Jonsson G, Bendahl PO, Holm K, et al. Identification of subtypes in human epidermal growth factor receptor 2–positive breast cancer reveals a gene signature prognostic of outcome. J Clin Oncol. 2010;28:1813–20. doi: 10.1200/JCO.2009.22.8775.
    1. Jonsson G, Staaf J, Vallon-Christersson J, Ringner M, Gruvberger-Saal SK, Saal LH, et al. The retinoblastoma gene undergoes rearrangements in BRCA1-deficient basal-like breast cancer. Cancer Res. 2012;72:4028–36. doi: 10.1158/0008-5472.CAN-12-0097.
    1. Sweden Cancerome Analysis Network - Breast. Available at: .
    1. Alkner S, Bendahl PO, Ferno M, Manjer J, Ryden L. Prediction of outcome after diagnosis of metachronous contralateral breast cancer. BMC Cancer. 2011;11:114. doi: 10.1186/1471-2407-11-114.
    1. Lund University. Faculty of Medicine - Oncology and Pathology. Available at: .
    1. Saal LH, Troein C, Vallon-Christersson J, Gruvberger S, Borg A, Peterson C. BioArray Software Environment (BASE): a platform for comprehensive management and analysis of microarray data. Genome Biol. 2002;3:SOFTWARE0003.
    1. Troein C, Vallon-Christersson J, Saal LH. An introduction to BioArray Software Environment. Methods Enzymol. 2006;411:99–119. doi: 10.1016/S0076-6879(06)11007-1.
    1. Vallon-Christersson J, Nordborg N, Svensson M, Hakkinen J. BASE–2nd generation software for microarray data management and analysis. BMC Bioinformatics. 2009;10:330. doi: 10.1186/1471-2105-10-330.
    1. Parkhomchuk D, Borodina T, Amstislavskiy V, Banaru M, Hallen L, Krobitsch S, et al. Transcriptome analysis by strand-specific sequencing of complementary DNA. Nucleic Acids Res. 2009;37:e123. doi: 10.1093/nar/gkp596.
    1. Nalpas NC, Park SD, Magee DA, Taraktsoglou M, Browne JA, Conlon KM, et al. Whole-transcriptome, high-throughput RNA sequence analysis of the bovine macrophage response to Mycobacterium bovis infection in vitro. BMC Genomics. 2013;14:230. doi: 10.1186/1471-2164-14-230.
    1. Borgstrom E, Lundin S, Lundeberg J. Large scale library generation for high throughput sequencing. PLoS One. 2011;6:e19119. doi: 10.1371/journal.pone.0019119.
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9:357–9. doi: 10.1038/nmeth.1923.
    1. Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R, Salzberg SL. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 2013;14:R36. doi: 10.1186/gb-2013-14-4-r36.
    1. Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc. 2012;7:562–78. doi: 10.1038/nprot.2012.016.
    1. Brueffer C. TopHat Recondition. Python script. Available at: .
    1. Morgan M, Pages H. Rsamtools: Binary alignment (BAM), variant call (BCF), or tabix file import. R package version 1.12.4. Available at: .
    1. Sørlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A. 2003;100:8418–23. doi: 10.1073/pnas.0932692100.
    1. Hu Z, Fan C, Oh DS, Marron JS, He X, Qaqish BF, et al. The molecular portraits of breast tumors are conserved across microarray platforms. BMC Genomics. 2006;7:96. doi: 10.1186/1471-2164-7-96.
    1. Parker JS, Mullins M, Cheang MC, Leung S, Voduc D, Vickery T, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol. 2009;27:1160–7. doi: 10.1200/JCO.2008.18.1370.
    1. Allen JD, Wang S, Chen M, Girard L, Minna JD, Xie Y, et al. Probe mapping across multiple microarray platforms. Brief Bioinform. 2012;13:547–54. doi: 10.1093/bib/bbr076.
    1. Atlas TCG. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70. doi: 10.1038/nature11412.
    1. Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, et al. The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012;486:400–4.
    1. Futreal PA, Coin L, Marshall M, Down T, Hubbard T, Wooster R, et al. A census of human cancer genes. Nat Rev Cancer. 2004;4:177–83. doi: 10.1038/nrc1299.
    1. bam-readcount. Available at: .
    1. Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–76. doi: 10.1101/gr.129684.111.
    1. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26:841–2. doi: 10.1093/bioinformatics/btq033.
    1. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38:e164. doi: 10.1093/nar/gkq603.
    1. The Cancer Genome Atlas. Available at: .
    1. Gene Expression Omnibus. Available at: .
    1. Saal LH, Holm K, Maurer M, Memeo L, Su T, Wang X, et al. PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast carcinoma. Cancer Res. 2005;65:2554–9. doi: 10.1158/0008-5472-CAN-04-3913.
    1. Budhu A, Forgues M, Ye QH, Jia HL, He P, Zanetti KA, et al. Prediction of venous metastases, recurrence, and prognosis in hepatocellular carcinoma based on a unique immune response signature of the liver microenvironment. Cancer Cell. 2006;10:99–111. doi: 10.1016/j.ccr.2006.06.016.
    1. Jais JP, Haioun C, Molina TJ, Rickman DS, de Reynies A, Berger F, et al. The expression of 16 genes related to the cell of origin and immune response predicts survival in elderly patients with diffuse large B-cell lymphoma treated with CHOP and rituximab. Leukemia. 2008;22:1917–24. doi: 10.1038/leu.2008.188.
    1. Roepman P, Jassem J, Smit EF, Muley T, Niklinski J, van de Velde T, et al. An immune response enriched 72-gene prognostic profile for early-stage non-small-cell lung cancer. Clin Cancer Res. 2009;15:284–90. doi: 10.1158/1078-0432.CCR-08-1258.
    1. Criscitiello C, Azim Jr HA, Schouten PC, Linn SC, Sotiriou C. Understanding the biology of triple-negative breast cancer. Ann Oncol. 2012;23:vi13–18.
    1. Shah SP, Roth A, Goya R, Oloumi A, Ha G, Zhao Y, et al. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature. 2012;486:395–9.
    1. Tang X, Baheti S, Shameer K, Thompson KJ, Wills Q, Niu N, et al. The eSNV-detect: a computational system to identify expressed single nucleotide variants from transcriptome sequencing data. Nucleic Acids Res. 2014;42:e172. doi: 10.1093/nar/gku1005.
    1. Wilkerson MD, Cabanski CR, Sun W, Hoadley KA, Walter V, Mose LE, et al. Integrated RNA and DNA sequencing improves mutation detection in low purity tumors. Nucleic Acids Res. 2014;42:e107. doi: 10.1093/nar/gku489.

Source: PubMed

3
Abonnere