RNA sequencing-based single sample predictors of molecular subtype and risk of recurrence for clinical assessment of early-stage breast cancer

Johan Staaf, Jari Häkkinen, Cecilia Hegardt, Lao H Saal, Siker Kimbung, Ingrid Hedenfalk, Tonje Lien, Therese Sørlie, Bjørn Naume, Hege Russnes, Rachel Marcone, Ayyakkannu Ayyanan, Cathrin Brisken, Rebecka R Malterling, Bengt Asking, Helena Olofsson, Henrik Lindman, Pär-Ola Bendahl, Anna Ehinger, Christer Larsson, Niklas Loman, Lisa Rydén, Martin Malmberg, Åke Borg, Johan Vallon-Christersson, Johan Staaf, Jari Häkkinen, Cecilia Hegardt, Lao H Saal, Siker Kimbung, Ingrid Hedenfalk, Tonje Lien, Therese Sørlie, Bjørn Naume, Hege Russnes, Rachel Marcone, Ayyakkannu Ayyanan, Cathrin Brisken, Rebecka R Malterling, Bengt Asking, Helena Olofsson, Henrik Lindman, Pär-Ola Bendahl, Anna Ehinger, Christer Larsson, Niklas Loman, Lisa Rydén, Martin Malmberg, Åke Borg, Johan Vallon-Christersson

Abstract

Multigene assays for molecular subtypes and biomarkers can aid management of early invasive breast cancer. Using RNA-sequencing we aimed to develop single-sample predictor (SSP) models for clinical markers, subtypes, and risk of recurrence (ROR). A cohort of 7743 patients was divided into training and test set. We trained SSPs for subtypes and ROR assigned by nearest-centroid (NC) methods and SSPs for biomarkers from histopathology. Classifications were compared with Prosigna in two external cohorts (ABiM, n = 100 and OSLO2-EMIT0, n = 103). Prognostic value was assessed using distant recurrence-free interval. Agreement between SSP and NC for PAM50 (five subtypes) was high (85%, Kappa = 0.78) for Subtype (four subtypes) very high (90%, Kappa = 0.84) and for ROR risk category high (84%, Kappa = 0.75, weighted Kappa = 0.90). Prognostic value was assessed as equivalent and clinically relevant. Agreement with histopathology was very high or high for receptor status, while moderate for Ki67 status and poor for Nottingham histological grade. SSP and Prosigna concordance was high for subtype (OSLO-EMIT0 83%, Kappa = 0.73 and ABiM 80%, Kappa = 0.72) and moderate and high for ROR risk category (68 and 84%, Kappa = 0.50 and 0.70, weighted Kappa = 0.70 and 0.78). Pooled concordance for emulated treatment recommendation dichotomized for chemotherapy was high (85%, Kappa = 0.66). Retrospective evaluation suggested that SSP application could change chemotherapy recommendations for up to 17% of postmenopausal ER+/HER2-/N0 patients with balanced escalation and de-escalation. Results suggest that NC and SSP models are interchangeable on a group-level and nearly so on a patient level and that SSP models can be derived to closely match clinical tests.

Conflict of interest statement

All authors declare no competing financial interests related to the study, and for all but A.E., H.L., and L.H.S. also no competing non-financial interests. A.E., H.L., and L.H.S. declare the following competing non-financial interests for this study: A.E. has received speakers’ honoraria from Novartis, Amgen, Roche, and advisory board fees from Roche. H.L. reports honoraria for lecturing from Astra-Zeneca, Novartis, Lilly, and Seagen, and working in advisory boards of Pfizer, Novartis, Daiichi, MSD, Amgen, and Pierre Fabre, and has received research support from Roche. L.H.S. has employment and ownership interest (including stock and patents) in SAGA Diagnostics AB.

© 2022. The Author(s).

Figures

Fig. 1. Outline of the study.
Fig. 1. Outline of the study.
a Study cohorts: SCAN-B, ABiM, and OSLO2-EMIT0. Available data types and usage outlined. FT fresh-frozen tissue, FFPE formalin-fixed paraffin-embedded tissue. b Scheme for development and validation of SSP models for molecular subtypes and ROR versus NCN equivalents. Scheme outlines created SSP models and their usage in different cohorts. NCN and SSP 4-class subtype models include Basal-like, HER2-enriched, Luminal A and Luminal B subtypes. Binary SSP-ROR and NCN-ROR risk classes were created similar as described by Bartlett et al.. An emulated 3-group SSP-based treatment recommendation (SSP ROR ETR) was created based on published Norwegian guidelines for Prosigna usage and applied to relevant SCAN-B patients based on guidelines. c Scheme for development and validation of SSPs for clinical markers: ER, PR, HER2, Ki67, and NHG. Scheme outlines created SSP models and their usage in different cohorts.
Fig. 2. Validation of SSP classifications against…
Fig. 2. Validation of SSP classifications against NCN classifications in the independent test set of early breast cancer.
a Agreement chart and confusion matrix comparing SSP classifications (x-axis/columns) with NCN classifications (y-axis/rows) for PAM50 (five subtypes) and b Subtype (four subtypes). c Scatterplot of binned NCN-ROR values (y-axis) versus SSP-ROR (x-axis). d Boxplot of NCN-ROR values (y-axis) by SSP-ROR (x-axis). e Distributions of NCN-ROR values and f SSP-ROR values by SSP-PAM50 (five subtypes). g Distributions of NCN-ROR values or h SSP-ROR values, by SSP-Subtype (four subtypes) for ER+/HER2- breast cancer classified as Luminal A or Luminal B. i Agreement chart and confusion matrix comparing SSP classification (x-axis/columns) with NCN classification (y-axis/rows) for ROR risk classification and j emulated treatment recommendation. Boxplot elements: center line (median), bounds of box (upper and lower quartiles), whiskers (1.5x interquartile range).
Fig. 3. Validation of SSP models for…
Fig. 3. Validation of SSP models for clinical markers in the independent population-based test set of early breast cancer.
Agreement chart and confusion matrix comparing the SSP classifications (x-axis/columns) with clinical histopathology status (y-axis/rows) for a ER status, b PR status, c HER2 status using a general SSP model, d HER2 status using a SSP model specific for ER status, e Ki67-status, f NHG.
Fig. 4. Assessment of prognostic value of…
Fig. 4. Assessment of prognostic value of SSP stratification and NCN stratification.
Comparison of SSP and NCN classifications in the independent population-based test set by assessment of prognostic value. Kaplan-Meier plots for molecular subtype with five groups (PAM50) or four groups (Subtype) using DRFi as clinical endpoint: a PAM50 by NCN (left) and SSP (right), b Subtype by NCN (left) and SSP (right). c Cox regression analysis using DRFi as endpoint in the test set restricted to patients with ER+/HER2-/N0 disease diagnosed over 50 years of age that only received endocrine adjuvant treatment (n = 772). Test and reference group is specified on the left. Hazard ratios and 95% confidence interval ranges from univariable analysis (left forest plot) and multivariable analysis (right forest plot) with tumor size, age at diagnosis, and NHG as covariates. Kaplan–Meier plots for stratification of ER+/HER2-/N0 disease diagnosed over 50 that only received endocrine adjuvant treatment in the test set by NCN (left in each panel) and SSP (right in each panel) for: d PAM50 subtype, e Subtype, f ROR risk classification, and g the two-group ROR stratification according to Bartlett et al.. Error bars correspond to 95% confidence interval.
Fig. 5. SSP classifications for Subtype and…
Fig. 5. SSP classifications for Subtype and ROR risk category in early-stage breast cancer and cross-comparison with administered systemic treatment.
The basis for comparisons is the 6660-patient follow-up cohort (Table 1). Summarized proportions are shown on the right side of bar graphs. The first and last year of enrollment (2010 and 2018) are not full calendar years and therefore include notably smaller numbers of enrolled patients. a Proportions of SSP-Subtype by year of diagnosis. b Proportions of SSP-ROR risk category by year of diagnosis. c Proportions for adjuvant treatment within ER+/HER2-/N0 patients diagnosed at age >50 years by different age at diagnosis. Endo: endocrine therapy only, ChemoEndo: adjuvant chemotherapy and endocrine therapy. None: no adjuvant systemic therapy. d Proportions for adjuvant treatment within ER+/HER2-/N0 patients diagnosed at age >50 ≤ 70 years by year of diagnosis. e Cross-comparison of the naive SSP ETR dichotomized for chemotherapy (yes/no) with records of administered systemic treatment within ER+/HER2-/N0 patients at age >50 ≤ 70. The groups for which SSP treatment recommendation is in agreement with the administered treatment are shown in black for regimen without chemotherapy (No CT) and in red for regimen including chemotherapy (CT). The discordant groups where SSP would lead to escalation of treatment (No CT to CT) are shown in orange and de-escalation of treatment (CT to No CT) in blue. f Kaplan-Meier plot for SSP stratification by SSP-ETR treatment recommendation within the N0 subgroup of ER + /HER2- patients diagnosed at age >50 ≤ 70 and no adjuvant treatment. g Kaplan-Meier plot for SSP stratification by SSP-ETR dichotomized for chemotherapy (chemotherapy vs. no chemotherapy) within ER+/HER2-/N0 patients diagnosed at age >50 ≤ 70 treated with adjuvant endocrine therapy only. h Forest plots of Hazard ratios and 95% confidence interval ranges from univariable and i multivariable Cox regression using DRFi as endpoint stratified using SSP treatment recommendation. Multivariable analysis is with tumor size, age at diagnosis, and NHG as covariates. Test (SSP stratification) and subgroup (administered treatment) is specified on the left of the univariable forest plot. Error bars correspond to 95% confidence interval.

References

    1. Goldhirsch A, et al. 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. doi: 10.1093/annonc/mdt303.
    1. Cardoso F, et al. 70-Gene signature as an aid to treatment decisions in early-stage breast cancer. N. Engl. J. Med. 2016;375:717–729. doi: 10.1056/NEJMoa1602253.
    1. Gnant M, et al. Predicting distant recurrence in receptor-positive breast cancer patients with limited clinicopathological risk: using the PAM50 Risk of Recurrence score in 1478 postmenopausal patients of the ABCSG-8 trial treated with adjuvant endocrine therapy alone. Ann. Oncol. 2014;25:339–345. doi: 10.1093/annonc/mdt494.
    1. Sparano JA, et al. Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N. Engl. J. Med. 2018;379:111–121. doi: 10.1056/NEJMoa1804710.
    1. Bartlett JM, et al. Comparing breast cancer multiparameter tests in the OPTIMA prelim trial: no test is more equal than the others. J. Natl Cancer Inst. 2016;108:djw050. doi: 10.1093/jnci/djw050.
    1. Andre F, et al. Use of biomarkers to guide decisions on adjuvant systemic therapy for women with early-stage invasive breast cancer: ASCO clinical practice guideline update-integration of results from TAILORx. J. Clin. Oncol. 2019;37:1956–1964. doi: 10.1200/JCO.19.00945.
    1. Parker JS, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J. Clin. Oncol. 2009;27:1160–1167. doi: 10.1200/JCO.2008.18.1370.
    1. Wallden B, et al. Development and verification of the PAM50-based Prosigna breast cancer gene signature assay. BMC Med. Genomics. 2015;8:54. doi: 10.1186/s12920-015-0129-6.
    1. Laenkholm AV, et al. Population-based study of Prosigna-PAM50 and outcome among postmenopausal women with estrogen receptor-positive and HER2-negative operable invasive lobular or ductal breast cancer. Clin. Breast Cancer. 2020;20:e423–e432. doi: 10.1016/j.clbc.2020.01.013.
    1. Laenkholm AV, et al. PAM50 risk of recurrence score predicts 10-year distant recurrence in a comprehensive danish cohort of postmenopausal women allocated to 5 years of endocrine therapy for hormone receptor-positive early breast cancer. J. Clin. Oncol. 2018;36:735–740. doi: 10.1200/JCO.2017.74.6586.
    1. Gnant M, et al. Identifying clinically relevant prognostic subgroups of postmenopausal women with node-positive hormone receptor-positive early-stage breast cancer treated with endocrine therapy: a combined analysis of ABCSG-8 and ATAC using the PAM50 risk of recurrence score and intrinsic subtype. Ann. Oncol. 2015;26:1685–1691. doi: 10.1093/annonc/mdv215.
    1. Ohnstad HO, et al. Prognostic value of PAM50 and risk of recurrence score in patients with early-stage breast cancer with long-term follow-up. Breast Cancer Res. 2017;19:120. doi: 10.1186/s13058-017-0911-9.
    1. Picornell AC, et al. Breast cancer PAM50 signature: correlation and concordance between RNA-Seq and digital multiplexed gene expression technologies in a triple negative breast cancer series. BMC Genomics. 2019;20:452. doi: 10.1186/s12864-019-5849-0.
    1. Paquet ER, Hallett MT. Absolute assignment of breast cancer intrinsic molecular subtype. J. Natl Cancer Inst. 2015;107:357. doi: 10.1093/jnci/dju357.
    1. Sorlie T, et al. The importance of gene-centring microarray data. Lancet Oncol. 2010;11:719–720. doi: 10.1016/S1470-2045(10)70174-1.
    1. Staaf J, Ringner M. Making breast cancer molecular subtypes robust? J. Natl Cancer Inst. 2015;107:386. doi: 10.1093/jnci/dju386.
    1. Ringner M, Jonsson G, Staaf J. Prognostic and chemotherapy predictive value of gene-expression phenotypes in primary lung adenocarcinoma. Clin. Cancer Res. 2016;22:218–229. doi: 10.1158/1078-0432.CCR-15-0529.
    1. Prat A, Parker JS. Standardized versus research-based PAM50 intrinsic subtyping of breast cancer. Clin. Transl. Oncol. 2020;22:953–955. doi: 10.1007/s12094-019-02203-x.
    1. Afsari B, Fertig EJ, Geman D, Marchionni L. switchBox: an R package for k-Top Scoring Pairs classifier development. Bioinformatics. 2015;31:273–274. doi: 10.1093/bioinformatics/btu622.
    1. Cirenajwis H, Lauss M, Planck M, Vallon-Christersson J, Staaf J. Performance of gene expression-based single sample predictors for assessment of clinicopathological subgroups and molecular subtypes in cancers: a case comparison study in non-small cell lung cancer. Brief. Bioinform. 2020;21:729–740. doi: 10.1093/bib/bbz008.
    1. Saal LH, 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. doi: 10.1186/s13073-015-0131-9.
    1. Vallon-Christersson J, et al. Cross comparison and prognostic assessment of breast cancer multigene signatures in a large population-based contemporary clinical series. Sci. Rep. 2019;9:12184. doi: 10.1038/s41598-019-48570-x.
    1. Ryden L, et al. Minimizing inequality in access to precision medicine in breast cancer by real-time population-based molecular analysis in the SCAN-B initiative. Br. J. Surg. 2018;105:e158–e168. doi: 10.1002/bjs.10741.
    1. Nielsen T, et al. Analytical validation of the PAM50-based Prosigna Breast Cancer Prognostic Gene Signature Assay and nCounter Analysis System using formalin-fixed paraffin-embedded breast tumor specimens. BMC Cancer. 2014;14:177. doi: 10.1186/1471-2407-14-177.
    1. Sotiriou C, et al. Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J. Natl Cancer Inst. 2006;98:262–272. doi: 10.1093/jnci/djj052.
    1. Ivshina AV, et al. Genetic reclassification of histologic grade delineates new clinical subtypes of breast cancer. Cancer Res. 2006;66:10292–10301. doi: 10.1158/0008-5472.CAN-05-4414.
    1. Lofgren L, et al. Validation of data quality in the Swedish National Register for Breast Cancer. BMC Public Health. 2019;19:495. doi: 10.1186/s12889-019-6846-6.
    1. Staaf J, et al. Whole-genome sequencing of triple-negative breast cancers in a population-based clinical study. Nat. Med. 2019;25:1526–1533. doi: 10.1038/s41591-019-0582-4.
    1. Brueffer, C. et al. Clinical value of RNA sequencing-based classifiers for prediction of the five conventional breast cancer biomarkers: a report from the population-based multicenter Sweden Cancerome Analysis Network-breast initiative. JCO Precis Oncol.2, PO.17.00135 (2018).
    1. Sorlie T, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc. Natl Acad. Sci. USA. 2003;100:8418–8423. doi: 10.1073/pnas.0932692100.
    1. Lien TG, et al. Sample preparation approach influences PAM50 risk of recurrence score in early breast cancer. Cancers. 2021;13:6118. doi: 10.3390/cancers13236118.
    1. Rantalainen M, et al. Sequencing-based breast cancer diagnostics as an alternative to routine biomarkers. Sci. Rep. 2016;6:38037. doi: 10.1038/srep38037.
    1. Bastani M, et al. A machine learned classifier that uses gene expression data to accurately predict estrogen receptor status. PLoS ONE. 2013;8:e82144. doi: 10.1371/journal.pone.0082144.
    1. National Quality Registry for breast cancer - Sweden. (2021).
    1. Stalhammar G, et al. Digital image analysis of Ki67 in hot spots is superior to both manual Ki67 and mitotic counts in breast cancer. Histopathology. 2018;72:974–989. doi: 10.1111/his.13452.
    1. Acs, B. et al. Variability in breast cancer biomarker assessment and the effect on oncological treatment decisions: a nationwide 5-year population-based study. Cancers13, 1166 (2021).
    1. Nielsen TO, et al. Assessment of Ki67 in breast cancer: updated recommendations from the international Ki67 in breast cancer working group. J. Natl Cancer Inst. 2021;113:808–819. doi: 10.1093/jnci/djaa201.
    1. Desmedt C, et al. Biological processes associated with breast cancer clinical outcome depend on the molecular subtypes. Clin. Cancer Res. 2008;14:5158–5165. doi: 10.1158/1078-0432.CCR-07-4756.
    1. Mackay A, et al. Microarray-based class discovery for molecular classification of breast cancer: analysis of interobserver agreement. J. Natl Cancer Inst. 2011;103:662–673. doi: 10.1093/jnci/djr071.
    1. Fan C, et al. Concordance among gene-expression-based predictors for breast cancer. N. Engl. J. Med. 2006;355:560–569. doi: 10.1056/NEJMoa052933.
    1. Buus R, et al. Molecular drivers of oncotype DX, Prosigna, EndoPredict, and the Breast Cancer Index: A TransATAC study. J. Clin. Oncol. 2021;39:126–135. doi: 10.1200/JCO.20.00853.
    1. Ontario H. Gene expression profiling tests for early-stage invasive breast cancer: a health technology assessment. Ont. Health Technol. Assess. Ser. 2020;20:1–234.
    1. Brueffer, C. et al. The mutational landscape of the SCAN-B real-world primary breast cancer transcriptome. EMBO Mol. Med.12, e12118 (2020).
    1. Dahlgren M, et al. Preexisting somatic mutations of estrogen receptor alpha (ESR1) in early-stage primary breast cancer. JNCI Cancer Spectr. 2021;5:pkab028. doi: 10.1093/jncics/pkab028.
    1. Shamseddin, M. et al. Contraceptive progestins with androgenic properties stimulate breast epithelial cell proliferation. EMBO Mol. Med.13, e14314 (2021).
    1. Swedish Society of Pathology. (2021).
    1. Aure MR, et al. Integrative clustering reveals a novel split in the luminal A subtype of breast cancer with impact on outcome. Breast Cancer Res. 2017;19:44. doi: 10.1186/s13058-017-0812-y.
    1. Winter C, et al. Targeted sequencing of BRCA1 and BRCA2 across a large unselected breast cancer cohort suggests that one-third of mutations are somatic. Ann. Oncol. 2016;27:1532–1538. doi: 10.1093/annonc/mdw209.
    1. Tekpli X, et al. An independent poor-prognosis subtype of breast cancer defined by a distinct tumor immune microenvironment. Nat. Commun. 2019;10:5499. doi: 10.1038/s41467-019-13329-5.
    1. Picard tools. (2021).
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30:2114–2120. doi: 10.1093/bioinformatics/btu170.
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat. Methods. 2012;9:357–359. doi: 10.1038/nmeth.1923.
    1. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods. 2015;12:357–360. doi: 10.1038/nmeth.3317.
    1. Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 2019;37:907–915. doi: 10.1038/s41587-019-0201-4.
    1. Pertea M, et al. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 2015;33:290–295. doi: 10.1038/nbt.3122.
    1. dbSNP. (2021).
    1. Frankish A, et al. GENCODE reference annotation for the human and mouse genomes. Nucleic Acids Res. 2019;47:D766–D773. doi: 10.1093/nar/gky955.
    1. AIMS GitHub repository. (2021).
    1. Gourgou-Bourgade S, et al. Guidelines for time-to-event end point definitions in breast cancer trials: results of the DATECAN initiative (Definition for the Assessment of Time-to-event Endpoints in CANcer trials) dagger. Ann. Oncol. 2015;26:873–879. doi: 10.1093/annonc/mdv106.
    1. Brooke HL, et al. The Swedish cause of death register. Eur. J. Epidemiol. 2017;32:765–773. doi: 10.1007/s10654-017-0316-1.
    1. Vallon-Christersson, J. RNA Sequencing-based single sample predictors of molecular subtype and risk of recurrence for clinical assessment of early-stage breast cancer. Mendeley DataV1. (10.17632/yzxtxn4nmd.1) (2022).

Source: PubMed

3
Abonnieren