Minimizing inequality in access to precision medicine in breast cancer by real-time population-based molecular analysis in the SCAN-B initiative

L Rydén, N Loman, C Larsson, C Hegardt, J Vallon-Christersson, M Malmberg, H Lindman, A Ehinger, L H Saal, Å Borg, L Rydén, N Loman, C Larsson, C Hegardt, J Vallon-Christersson, M Malmberg, H Lindman, A Ehinger, L H Saal, Å Borg

Abstract

Background: Selection of systemic therapy for primary breast cancer is currently based on clinical biomarkers along with stage. Novel genomic tests are continuously being introduced as more precise tools for guidance of therapy, although they are often developed for specific patient subgroups. The Sweden Cancerome Analysis Network - Breast (SCAN-B) initiative aims to include all patients with breast cancer for tumour genomic analysis, and to deliver molecular subtype and mutational data back to the treating physician.

Methods: An infrastructure for collection of blood and fresh tumour tissue from all patients newly diagnosed with breast cancer was set up in 2010, initially including seven hospitals within the southern Sweden regional catchment area, which has 1.8 million inhabitants. Inclusion of patients was implemented into routine clinical care, with collection of tumour tissue at local pathology departments for transport to the central laboratory, where routines for rapid sample processing, RNA sequencing and biomarker reporting were developed.

Results: More than 10 000 patients from nine hospitals have currently consented to inclusion in SCAN-B with high (90 per cent) inclusion rates from both university and secondary hospitals. Tumour samples and successful RNA sequencing are being obtained from more than 70 per cent of patients, showing excellent representation compared with the national quality registry as a truly population-based cohort. Molecular biomarker reports can be delivered to multidisciplinary conferences within 1 week.

Conclusion: Population-based collection of fresh tumour tissue is feasible given a decisive joint effort between academia and collaborative healthcare groups, and with governmental support. An infrastructure for genomic analysis and prompt data output paves the way for novel systemic therapy for patients from all hospitals, irrespective of size and location.

© 2018 The Authors. BJS published by John Wiley & Sons Ltd on behalf of BJS Society Ltd.

Figures

Figure 1
Figure 1
Infrastructure of the SCAN‐B study and the catchment area
Figure 2
Figure 2
SCAN‐B enrolment statistics, and representativity of enrolled patients and sampled tumours. a Number of enrolled patients with operable invasive primary breast cancer by calendar year for 2011–2016 (partial years 2010 and 2017 not shown) and number of cases for which a preoperative untreated tumour specimen was received and processed for analysis. b Proportion of all operable invasive primary breast cancer diagnoses in the catchment area that were not accrued, accrued but without a tumour specimen or accrued with a specimen. The statistics are restricted to the original seven hospital sites where enrolment was fully operational by early 2010. c–h Bar graphs illustrating the distribution of clinicopathological characteristics for all operable invasive primary breast cancer diagnoses, all patients accrued in SCAN‐B, and all patients accrued and with a processed tumour specimen: oestrogen receptor status (c), Nottingham histological grade (d), tumour size (e), age at diagnosis (f), lymph node status (g) and type of breast surgery (h)
Figure 3
Figure 3
Schematic illustration of the work flow from inclusion and surgical excision to output data via RNA/DNA extraction. ER, oestrogen receptor; PR, progesterone receptor, HER2, human epidermal growth factor receptor 2

References

    1. Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA et al Molecular portraits of human breast tumours. Nature 2000; 406: 747–52.1.
    1. Early Breast Cancer Trialists' Collaborative Group (EBCTCG) , Davies C, Godwin J, Gray R, Clarke M, Cutter D, Darby S et al Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient‐level meta‐analysis of randomised trials. Lancet 2011; 378: 771–784.
    1. Early Breast Cancer Trialists' Collaborative Group (EBCTCG) , Peto R, Davies C, Godwin J, Gray R, Pan HC, Clarke M et al Comparisons between different polychemotherapy regimens for early breast cancer: meta‐analyses of long‐term outcome among 100 000 women in 123 randomised trials. Lancet 2012; 379: 432–444.
    1. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER‐2/neu oncogene. Science 1987; 235: 177–182.
    1. Moja L, Tagliabue L, Balduzzi S, Parmelli E, Pistotti V, Guarneri V et al Trastuzumab containing regimens for early breast cancer. Cochrane Database Syst Rev 2012; (4)CD006243.
    1. Balduzzi S, Mantarro S, Guarneri V, Tagliabue L, Pistotti V, Moja L et al Trastuzumab‐containing regimens for metastatic breast cancer. Cochrane Database Syst Rev 2014; (6)CD006242.
    1. Swain SM, Kim SB, Cortés J, Ro J, Semiglazov V, Campone M et al Pertuzumab, trastuzumab, and docetaxel for HER2‐positive metastatic breast cancer (CLEOPATRA study): overall survival results from a randomised, double‐blind, placebo‐controlled, phase 3 study. Lancet Oncol 2013; 14: 461–471.
    1. Krop IE, Kim SB, Martin AG, LoRusso PM, Ferrero JM, Badovinac‐Crnjevic T et al Trastuzumab emtansine versus treatment of physician's choice in patients with previously treated HER2‐positive metastatic breast cancer (TH3RESA): final overall survival results from a randomised open‐label phase 3 trial. Lancet Oncol 2017; 18: 743–754.
    1. Blackwell KL, Burstein HJ, Storniolo AM, Rugo HS, Sledge G, Aktan G et al Overall survival benefit with lapatinib in combination with trastuzumab for patients with human epidermal growth factor receptor 2‐positive metastatic breast cancer: final results from the EGF104900 Study. J Clin Oncol 2012; 30: 2585–2592.
    1. Cameron D, Casey M, Oliva C, Newstat B, Imwalle B, Geyer CE. Lapatinib plus capecitabine in women with HER‐2‐positive advanced breast cancer: final survival analysis of a phase III randomized trial. Oncologist 2010; 15: 924–934.
    1. Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart‐Gebhart M, Thürlimann B et al; Panel members. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol 2013; 24: 2206–2223.
    1. Coates AS, Winer EP, Goldhirsch A, Gelber RD, Gnant M, Piccart‐Gebhart M et al Tailoring therapies – improving the management of early breast cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015. Ann Oncol 2015; 26: 1533–1546.
    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: 530–536.
    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: 2817–2826.
    1. Cardoso F, van't Veer LJ, Bogaerts J, Slaets L, Viale G, Delaloge S et al; MINDACT Investigators. 70‐gene signature as an aid to treatment decisions in early‐stage breast cancer. N Engl J Med 2016; 375: 717–729.
    1. Regionala cancercentrum i samverkan . Styrdokument för nationellt Kvalitetsregister bröstcancer inklusive bröstrekonstruktion. [accessed 3 September 2017].
    1. Socialstyrelsen . Nationella riktlinjer för bröst‐, prostata‐, tjocktarms‐ och ändtarmscancervård. [accessed 3 September 2017].
    1. Finch AP, Lubinski J, Møller P, Singer CF, Karlan B, Senter L et al Impact of oophorectomy on cancer incidence and mortality in women with a BRCA1 or BRCA2 mutation. J Clin Oncol 2014; 32: 1547 – 1553
    1. Ludwig KK, Neuner J, Butler A, Geurts JL, Kong AL. Risk reduction and survival benefit of prophylactic surgery in BRCA mutation carriers, a systematic review. Am J Surg 2016; 212: 660–669.
    1. Hayes DF, Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Miller MC et al Circulating tumor cells at each follow‐up time point during therapy of metastatic breast cancer patients predict progression‐free and overall survival. Clin Cancer Res 2006; 12: 4218–4224.
    1. Bidard FC, Peeters DJ, Fehm T, Nolé F, Gisbert‐Criado R, Mavroudis D et al Clinical validity of circulating tumour cells in patients with metastatic breast cancer: a pooled analysis of individual patient data. Lancet Oncol 2014; 15: 406–414.
    1. Olsson E, Winter C, George A, Chen Y, Howlin J, Tang MH et al Serial monitoring of circulating tumor DNA in patients with primary breast cancer for detection of occult metastatic disease. EMBO Mol Med 2015; 7: 1034–1047.
    1. Loman N, Saal LH. The state of the art in prediction of breast cancer relapse using cell‐free circulating tumor DNA liquid biopsies. Ann Transl Med 2016; 4(Suppl 1): S68.
    1. Sigurdsson H, Baldetorp B, Borg A, Dalberg M, Fernö M, Killander D et al Indicators of prognosis in node‐negative breast cancer. N Engl J Med 1990; 322: 1045–1053.
    1. Fernö M, Stål O, Baldetorp B, Hatschek T, Källström AC, Malmström P et al Results of two or five years of adjuvant tamoxifen correlated to steroid receptor and S‐phase levels. South Sweden Breast Cancer Group, and South‐East Sweden Breast Cancer Group. Breast Cancer Res Treat 2000; 59: 69–76.
    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: 539–548.
    1. Gruvberger S, Ringnér 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: 5979–5984.
    1. Gruvberger‐Saal SK, Edén P, Ringnér M, Baldetorp B, Chebil G, Borg A et al Predicting continuous values of prognostic markers in breast cancer from microarray gene expression profiles. Mol Cancer Ther 2004; 3: 161–168.
    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: 2554–2559.
    1. Jonsson G, Naylor TL, Vallon‐Christersson J, Staaf J, Huang J, Ward MR et al Distinct genomic profiles in hereditary breast tumors identified by array‐based comparative genomic hybridization. Cancer Res 2005; 65: 7612–7621.
    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: 7564–7569.
    1. Saal LH, Gruvberger‐Saal SK, Persson C, Lövgren K, Jumppanen M, Staaf J et al Recurrent gross mutations of the PTEN tumor suppressor gene in breast cancers with deficient DSB repair. Nat Genet 2008; 40: 102–107.
    1. Staaf J, Ringnér M, Vallon‐Christersson J, Jönsson 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: 1813–1820.
    1. Staaf J, Jönsson G, Ringnér M, Vallon‐Christersson J, Grabau D, Arason A et al High‐resolution genomic and expression analyses of copy number alterations in HER2‐amplified breast cancer. Breast Cancer Res 2010; 12: R25.
    1. Holm K, Hegardt C, Staaf J, Vallon‐Christersson J, Jönsson G, Olsson H et al Molecular subtypes of breast cancer are associated with characteristic DNA methylation patterns. Breast Cancer Res 2010; 12: R36.
    1. Jönsson G, Staaf J, Vallon‐Christersson J, Ringnér M, Holm K, Hegardt C et al Genomic subtypes of breast cancer identified by array‐comparative genomic hybridization display distinct molecular and clinical characteristics. Breast Cancer Res 2010; 12: R42.
    1. Saal LH, Vallon‐Christersson J, Häkkinen J, Hegardt C, Grabau D, Winter C et al 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. Genome Med 2015; 7: 20.
    1. Regeringskanslitet . En nationell cancerstrategi för framtiden. [accessed 3 September 2017].
    1. Lindquist KE, Karlsson A, Levéen P, Brunnström H, Reuterswärd C, Holm K et al Clinical framework for next generation sequencing based analysis of treatment predictive mutations and multiplexed gene fusion detection in non‐small cell lung cancer. Oncotarget 2017; 8: 34796–34810.
    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.
    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.
    1. Hakkinen J, Nordborg N, Mansson O, Vallon‐Christersson J. Implementation of an open source software solution for laboratory information management and automated RNAseq data analysis in a large‐scale cancer genomics initiative using BASE with extension package Reggie. bioRxiv 2016. [accessed 2 September 2017].
    1. Nilsson MP. Treatment of BRCA1/2‐associated breast cancer and identification of mutation carriers among breast cancer patients. PhD thesis. Lund University; 2017.
    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: 1160–1167.
    1. Easton DF, Pharoah PD, Antoniou AC, Tischkowitz M, Tavtigian SV, Nathanson KL et al Gene‐panel sequencing and the prediction of breast‐cancer risk. N Engl J Med 2015; 372: 2243–2257.
    1. Nik‐Zainal S, Davies H, Staaf J, Ramakrishna M, Glodzik D, Zou X et al Landscape of somatic mutations in 560 breast cancer whole‐genome sequences. Nature 2016; 534: 47–54.

Source: PubMed

3
Abonnieren