Skip to main content
Top
Published in: Breast Cancer Research 1/2019

Open Access 01-12-2019 | Magnetic Resonance Imaging | Research article

Radiomic signatures with contrast-enhanced magnetic resonance imaging for the assessment of breast cancer receptor status and molecular subtypes: initial results

Authors: Doris Leithner, Joao V. Horvat, Maria Adele Marino, Blanca Bernard-Davila, Maxine S. Jochelson, R. Elena Ochoa-Albiztegui, Danny F. Martinez, Elizabeth A. Morris, Sunitha Thakur, Katja Pinker

Published in: Breast Cancer Research | Issue 1/2019

Login to get access

Abstract

Background

To evaluate the diagnostic performance of radiomic signatures extracted from contrast-enhanced magnetic resonance imaging (CE-MRI) for the assessment of breast cancer receptor status and molecular subtypes.

Methods

One hundred and forty-three patients with biopsy-proven breast cancer who underwent CE-MRI at 3 T were included in this IRB-approved HIPAA-compliant retrospective study. The training dataset comprised 91 patients (luminal A, n = 49; luminal B, n = 8; HER2-enriched, n = 11; triple negative, n = 23), while the validation dataset comprised 52 patients from a second institution (luminal A, n = 17; luminal B, n = 17; triple negative, n = 18). Radiomic analysis of manually segmented tumors included calculation of features derived from the first-order histogram (HIS), co-occurrence matrix (COM), run-length matrix (RLM), absolute gradient (GRA), autoregressive model (ARM), discrete Haar wavelet transform (WAV), and lesion geometry (GEO). Fisher, probability of error and average correlation (POE + ACC), and mutual information coefficients were used for feature selection. Linear discriminant analysis followed by k-nearest neighbor classification (with leave-one-out cross-validation) was used for pairwise radiomic-based separation of receptor status and molecular subtypes. Histopathology served as the standard of reference.

Results

In the training dataset, radiomic signatures yielded the following accuracies > 80%: luminal B vs. luminal A, 84.2% (mainly based on COM features); luminal B vs. triple negative, 83.9% (mainly based on GEO features); luminal B vs. all others, 89% (mainly based on COM features); and HER2-enriched vs. all others, 81.3% (mainly based on COM features). Radiomic signatures were successfully validated in the separate validation dataset for luminal A vs. luminal B (79.4%) and luminal B vs. triple negative (77.1%).

Conclusions

In this preliminary study, radiomic signatures with CE-MRI enable the assessment of breast cancer receptor status and molecular subtypes with high diagnostic accuracy. These results need to be confirmed in future larger studies.
Appendix
Available only for authorised users
Literature
1.
go back to reference Carey LA, Perou CM, Livasy CA, Dressler LG, Cowan D, Conway K, et al. Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA. 2006;295:2492–502.CrossRef Carey LA, Perou CM, Livasy CA, Dressler LG, Cowan D, Conway K, et al. Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA. 2006;295:2492–502.CrossRef
2.
go back to reference Huber KE, Carey LA, Wazer DE. Breast cancer molecular subtypes in patients with locally advanced disease: impact on prognosis, patterns of recurrence, and response to therapy. Semin Radiat Oncol. 2009;19:204–10.CrossRef Huber KE, Carey LA, Wazer DE. Breast cancer molecular subtypes in patients with locally advanced disease: impact on prognosis, patterns of recurrence, and response to therapy. Semin Radiat Oncol. 2009;19:204–10.CrossRef
3.
go back to reference Lam SW, Jimenez CR, Boven E. Breast cancer classification by proteomic technologies: current state of knowledge. Cancer Treat Rev. 2014;40:129–38.CrossRef Lam SW, Jimenez CR, Boven E. Breast cancer classification by proteomic technologies: current state of knowledge. Cancer Treat Rev. 2014;40:129–38.CrossRef
4.
go back to reference Wirapati P, Sotiriou C, Kunkel S, Farmer P, Pradervand S, Haibe-Kains B, et al. Meta-analysis of gene expression profiles in breast cancer: toward a unified understanding of breast cancer subtyping and prognosis signatures. Breast Cancer Res. 2008;10:R65.CrossRef Wirapati P, Sotiriou C, Kunkel S, Farmer P, Pradervand S, Haibe-Kains B, et al. Meta-analysis of gene expression profiles in breast cancer: toward a unified understanding of breast cancer subtyping and prognosis signatures. Breast Cancer Res. 2008;10:R65.CrossRef
5.
go back to reference Haynes B, Sarma A, Nangia-Makker P, Shekhar MP. Breast cancer complexity: implications of intratumoral heterogeneity in clinical management. Cancer Metastasis Rev. 2017;36:547–55.CrossRef Haynes B, Sarma A, Nangia-Makker P, Shekhar MP. Breast cancer complexity: implications of intratumoral heterogeneity in clinical management. Cancer Metastasis Rev. 2017;36:547–55.CrossRef
6.
go back to reference Zardavas D, Irrthum A, Swanton C, Piccart M. Clinical management of breast cancer heterogeneity. Nat Rev Clin Oncol. 2015;12:381–94.CrossRef Zardavas D, Irrthum A, Swanton C, Piccart M. Clinical management of breast cancer heterogeneity. Nat Rev Clin Oncol. 2015;12:381–94.CrossRef
7.
go back to reference Martelotto LG, Ng CK, Piscuoglio S, Weigelt B, Reis-Filho JS. Breast cancer intra-tumor heterogeneity. Breast Cancer Res. 2014;16:210.CrossRef Martelotto LG, Ng CK, Piscuoglio S, Weigelt B, Reis-Filho JS. Breast cancer intra-tumor heterogeneity. Breast Cancer Res. 2014;16:210.CrossRef
8.
go back to reference Cancer Genome Atlas N. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70.CrossRef Cancer Genome Atlas N. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70.CrossRef
9.
go back to reference Prat A, Perou CM. Deconstructing the molecular portraits of breast cancer. Mol Oncol. 2011;5:5–23.CrossRef Prat A, Perou CM. Deconstructing the molecular portraits of breast cancer. Mol Oncol. 2011;5:5–23.CrossRef
10.
go back to reference Prat A, Pineda E, Adamo B, Galvan P, Fernandez A, Gaba L, et al. Clinical implications of the intrinsic molecular subtypes of breast cancer. Breast. 2015;24(Suppl 2):S26–35.CrossRef Prat A, Pineda E, Adamo B, Galvan P, Fernandez A, Gaba L, et al. Clinical implications of the intrinsic molecular subtypes of breast cancer. Breast. 2015;24(Suppl 2):S26–35.CrossRef
11.
go back to reference Mazurowski MA, Zhang J, Grimm LJ, Yoon SC, Silber JI. Radiogenomic analysis of breast cancer: luminal B molecular subtype is associated with enhancement dynamics at MR imaging. Radiology. 2014;273:365–72.CrossRef Mazurowski MA, Zhang J, Grimm LJ, Yoon SC, Silber JI. Radiogenomic analysis of breast cancer: luminal B molecular subtype is associated with enhancement dynamics at MR imaging. Radiology. 2014;273:365–72.CrossRef
12.
go back to reference Tsoutsou PG, Vozenin MC, Durham AD, Bourhis J. How could breast cancer molecular features contribute to locoregional treatment decision making? Crit Rev Oncol Hematol. 2017;110:43–8.CrossRef Tsoutsou PG, Vozenin MC, Durham AD, Bourhis J. How could breast cancer molecular features contribute to locoregional treatment decision making? Crit Rev Oncol Hematol. 2017;110:43–8.CrossRef
13.
go back to reference Ahn HJ, Jung SJ, Kim TH, Oh MK, Yoon HK. Differences in clinical outcomes between luminal A and B type breast cancers according to the St. Gallen Consensus 2013. J Breast Cancer. 2015;18:149–59.CrossRef Ahn HJ, Jung SJ, Kim TH, Oh MK, Yoon HK. Differences in clinical outcomes between luminal A and B type breast cancers according to the St. Gallen Consensus 2013. J Breast Cancer. 2015;18:149–59.CrossRef
14.
go back to reference Senn HJ. St. Gallen consensus 2013: optimizing and personalizing primary curative therapy of breast cancer worldwide. Breast Care (Basel). 2013;8:101.CrossRef Senn HJ. St. Gallen consensus 2013: optimizing and personalizing primary curative therapy of breast cancer worldwide. Breast Care (Basel). 2013;8:101.CrossRef
15.
go back to reference Pisco AO, Huang S. Non-genetic cancer cell plasticity and therapy-induced stemness in tumour relapse: ‘What does not kill me strengthens me’. Br J Cancer. 2015;112:1725–32.CrossRef Pisco AO, Huang S. Non-genetic cancer cell plasticity and therapy-induced stemness in tumour relapse: ‘What does not kill me strengthens me’. Br J Cancer. 2015;112:1725–32.CrossRef
16.
go back to reference Grimm LJ, Zhang J, Baker JA, Soo MS, Johnson KS, Mazurowski MA. Relationships between MRI breast imaging-reporting and data system (BI-RADS) lexicon descriptors and breast cancer molecular subtypes: internal enhancement is associated with luminal B subtype. Breast J. 2017;23:579–82.CrossRef Grimm LJ, Zhang J, Baker JA, Soo MS, Johnson KS, Mazurowski MA. Relationships between MRI breast imaging-reporting and data system (BI-RADS) lexicon descriptors and breast cancer molecular subtypes: internal enhancement is associated with luminal B subtype. Breast J. 2017;23:579–82.CrossRef
17.
go back to reference Li H, Zhu Y, Burnside ES, Huang E, Drukker K, Hoadley KA, et al. Quantitative MRI radiomics in the prediction of molecular classifications of breast cancer subtypes in the TCGA/TCIA data set. NPJ Breast Cancer. 2016;2:16012. Li H, Zhu Y, Burnside ES, Huang E, Drukker K, Hoadley KA, et al. Quantitative MRI radiomics in the prediction of molecular classifications of breast cancer subtypes in the TCGA/TCIA data set. NPJ Breast Cancer. 2016;2:16012.
18.
go back to reference Waugh SA, Purdie CA, Jordan LB, Vinnicombe S, Lerski RA, Martin P, et al. Magnetic resonance imaging texture analysis classification of primary breast cancer. Eur Radiol. 2016;26:322–30.CrossRef Waugh SA, Purdie CA, Jordan LB, Vinnicombe S, Lerski RA, Martin P, et al. Magnetic resonance imaging texture analysis classification of primary breast cancer. Eur Radiol. 2016;26:322–30.CrossRef
19.
go back to reference Fan M, Li H, Wang S, Zheng B, Zhang J, Li L. Radiomic analysis reveals DCE-MRI features for prediction of molecular subtypes of breast cancer. PLoS One. 2017;12:e0171683.CrossRef Fan M, Li H, Wang S, Zheng B, Zhang J, Li L. Radiomic analysis reveals DCE-MRI features for prediction of molecular subtypes of breast cancer. PLoS One. 2017;12:e0171683.CrossRef
20.
go back to reference Wu J, Sun X, Wang J, Cui Y, Kato F, Shirato H, et al. Identifying relations between imaging phenotypes and molecular subtypes of breast cancer: model discovery and external validation. J Magn Reson Imaging. 2017;46:1017–27.CrossRef Wu J, Sun X, Wang J, Cui Y, Kato F, Shirato H, et al. Identifying relations between imaging phenotypes and molecular subtypes of breast cancer: model discovery and external validation. J Magn Reson Imaging. 2017;46:1017–27.CrossRef
21.
go back to reference Wang J, Kato F, Oyama-Manabe N, Li R, Cui Y, Tha KK, et al. Identifying triple-negative breast cancer using background parenchymal enhancement heterogeneity on dynamic contrast-enhanced MRI: a pilot radiomics study. PLoS One. 2015;10:e0143308.CrossRef Wang J, Kato F, Oyama-Manabe N, Li R, Cui Y, Tha KK, et al. Identifying triple-negative breast cancer using background parenchymal enhancement heterogeneity on dynamic contrast-enhanced MRI: a pilot radiomics study. PLoS One. 2015;10:e0143308.CrossRef
22.
go back to reference Mayerhoefer ME, Schima W, Trattnig S, Pinker K, Berger-Kulemann V, Ba-Ssalamah A. Texture-based classification of focal liver lesions on MRI at 3.0 Tesla: a feasibility study in cysts and hemangiomas. J Magn Reson Imaging. 2010;32:352–9.CrossRef Mayerhoefer ME, Schima W, Trattnig S, Pinker K, Berger-Kulemann V, Ba-Ssalamah A. Texture-based classification of focal liver lesions on MRI at 3.0 Tesla: a feasibility study in cysts and hemangiomas. J Magn Reson Imaging. 2010;32:352–9.CrossRef
23.
go back to reference Thakur SB, Durando M, Milans S, Cho GY, Gennaro L, Sutton EJ, et al. Apparent diffusion coefficient in estrogen receptor-positive and lymph node-negative invasive breast cancers at 3.0T DW-MRI: a potential predictor for an oncotype Dx test recurrence score. J Magn Reson Imaging. 2018;47:401–9.CrossRef Thakur SB, Durando M, Milans S, Cho GY, Gennaro L, Sutton EJ, et al. Apparent diffusion coefficient in estrogen receptor-positive and lymph node-negative invasive breast cancers at 3.0T DW-MRI: a potential predictor for an oncotype Dx test recurrence score. J Magn Reson Imaging. 2018;47:401–9.CrossRef
24.
go back to reference Collewet G, Strzelecki M, Mariette F. Influence of MRI acquisition protocols and image intensity normalization methods on texture classification. Magn Reson Imaging. 2004;22:81–91.CrossRef Collewet G, Strzelecki M, Mariette F. Influence of MRI acquisition protocols and image intensity normalization methods on texture classification. Magn Reson Imaging. 2004;22:81–91.CrossRef
25.
go back to reference Hu Y, Dennis TJ. Textured image segmentation by context enhanced clustering. IEE Proc Vis Image Signal Process. 1994;141:413–21.CrossRef Hu Y, Dennis TJ. Textured image segmentation by context enhanced clustering. IEE Proc Vis Image Signal Process. 1994;141:413–21.CrossRef
26.
go back to reference Kashyap R, Chellappa R. Estimation and choice of neighbors in spatial-interaction models of images. IEEE Trans Inf Theory. 1983;29:60–72.CrossRef Kashyap R, Chellappa R. Estimation and choice of neighbors in spatial-interaction models of images. IEEE Trans Inf Theory. 1983;29:60–72.CrossRef
27.
go back to reference Hajek M, Dezortova M, Materka A, Lerski RA. Texture analysis for magnetic resonance imaging. Prague: Med4Publishing; 2006. p. 234. Hajek M, Dezortova M, Materka A, Lerski RA. Texture analysis for magnetic resonance imaging. Prague: Med4Publishing; 2006. p. 234.
28.
go back to reference Fukunaga K. Introduction to statistical pattern recognition. 2nd ed. San Diego: Academic Press Professional, Inc; 1990. Fukunaga K. Introduction to statistical pattern recognition. 2nd ed. San Diego: Academic Press Professional, Inc; 1990.
29.
go back to reference Duda R, Hart PE, Stork DG. Pattern classification. 2nd ed. United States: Wiley; 2012. Duda R, Hart PE, Stork DG. Pattern classification. 2nd ed. United States: Wiley; 2012.
30.
go back to reference Pinker K, Moy L, Sutton EJ, Mann RM, Weber M, Thakur SB, et al. Diffusion-weighted imaging with apparent diffusion coefficient mapping for breast cancer detection as a stand-alone parameter: comparison with dynamic contrast-enhanced and multiparametric magnetic resonance imaging. Investig Radiol. 2018;53:587–95. Pinker K, Moy L, Sutton EJ, Mann RM, Weber M, Thakur SB, et al. Diffusion-weighted imaging with apparent diffusion coefficient mapping for breast cancer detection as a stand-alone parameter: comparison with dynamic contrast-enhanced and multiparametric magnetic resonance imaging. Investig Radiol. 2018;53:587–95.
31.
go back to reference Hon JD, Singh B, Sahin A, Du G, Wang J, Wang VY, et al. Breast cancer molecular subtypes: from TNBC to QNBC. Am J Cancer Res. 2016;6:1864–72.PubMedPubMedCentral Hon JD, Singh B, Sahin A, Du G, Wang J, Wang VY, et al. Breast cancer molecular subtypes: from TNBC to QNBC. Am J Cancer Res. 2016;6:1864–72.PubMedPubMedCentral
32.
go back to reference Sutton EJ, Dashevsky BZ, Oh JH, Veeraraghavan H, Apte AP, Thakur SB, et al. Breast cancer molecular subtype classifier that incorporates MRI features. J Magn Reson Imaging. 2016;44:122–9.CrossRef Sutton EJ, Dashevsky BZ, Oh JH, Veeraraghavan H, Apte AP, Thakur SB, et al. Breast cancer molecular subtype classifier that incorporates MRI features. J Magn Reson Imaging. 2016;44:122–9.CrossRef
33.
go back to reference Holli-Helenius K, Salminen A, Rinta-Kiikka I, Koskivuo I, Bruck N, Bostrom P, et al. MRI texture analysis in differentiating luminal A and luminal B breast cancer molecular subtypes - a feasibility study. BMC Med Imaging. 2017;17:69.CrossRef Holli-Helenius K, Salminen A, Rinta-Kiikka I, Koskivuo I, Bruck N, Bostrom P, et al. MRI texture analysis in differentiating luminal A and luminal B breast cancer molecular subtypes - a feasibility study. BMC Med Imaging. 2017;17:69.CrossRef
34.
go back to reference Mayerhoefer ME, Szomolanyi P, Jirak D, Materka A, Trattnig S. Effects of MRI acquisition parameter variations and protocol heterogeneity on the results of texture analysis and pattern discrimination: an application-oriented study. Med Phys. 2009;36:1236–43.CrossRef Mayerhoefer ME, Szomolanyi P, Jirak D, Materka A, Trattnig S. Effects of MRI acquisition parameter variations and protocol heterogeneity on the results of texture analysis and pattern discrimination: an application-oriented study. Med Phys. 2009;36:1236–43.CrossRef
35.
go back to reference Li Z, Mao Y, Li H, Yu G, Wan H, Li B. Differentiating brain metastases from different pathological types of lung cancers using texture analysis of T1 postcontrast MR. Magn Reson Med. 2016;76:1410–9.CrossRef Li Z, Mao Y, Li H, Yu G, Wan H, Li B. Differentiating brain metastases from different pathological types of lung cancers using texture analysis of T1 postcontrast MR. Magn Reson Med. 2016;76:1410–9.CrossRef
36.
go back to reference Fruehwald-Pallamar J, Czerny C, Holzer-Fruehwald L, Nemec SF, Mueller-Mang C, Weber M, et al. Texture-based and diffusion-weighted discrimination of parotid gland lesions on MR images at 3.0 Tesla. NMR Biomed. 2013;26:1372–9.CrossRef Fruehwald-Pallamar J, Czerny C, Holzer-Fruehwald L, Nemec SF, Mueller-Mang C, Weber M, et al. Texture-based and diffusion-weighted discrimination of parotid gland lesions on MR images at 3.0 Tesla. NMR Biomed. 2013;26:1372–9.CrossRef
Metadata
Title
Radiomic signatures with contrast-enhanced magnetic resonance imaging for the assessment of breast cancer receptor status and molecular subtypes: initial results
Authors
Doris Leithner
Joao V. Horvat
Maria Adele Marino
Blanca Bernard-Davila
Maxine S. Jochelson
R. Elena Ochoa-Albiztegui
Danny F. Martinez
Elizabeth A. Morris
Sunitha Thakur
Katja Pinker
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2019
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-019-1187-z

Other articles of this Issue 1/2019

Breast Cancer Research 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine