A novel representation of inter-site tumour heterogeneity from pre-treatment computed tomography textures classifies ovarian cancers by clinical outcome

Hebert Alberto Vargas, Harini Veeraraghavan, Maura Micco, Stephanie Nougaret, Yulia Lakhman, Andreas A Meier, Ramon Sosa, Robert A Soslow, Douglas A Levine, Britta Weigelt, Carol Aghajanian, Hedvig Hricak, Joseph Deasy, Alexandra Snyder, Evis Sala, Hebert Alberto Vargas, Harini Veeraraghavan, Maura Micco, Stephanie Nougaret, Yulia Lakhman, Andreas A Meier, Ramon Sosa, Robert A Soslow, Douglas A Levine, Britta Weigelt, Carol Aghajanian, Hedvig Hricak, Joseph Deasy, Alexandra Snyder, Evis Sala

Abstract

Purpose: To evaluate the associations between clinical outcomes and radiomics-derived inter-site spatial heterogeneity metrics across multiple metastatic lesions on CT in patients with high-grade serous ovarian cancer (HGSOC).

Methods: IRB-approved retrospective study of 38 HGSOC patients. All sites of suspected HGSOC involvement on preoperative CT were manually segmented. Gray-level correlation matrix-based textures were computed from each tumour site, and grouped into five clusters using a Gaussian Mixture Model. Pairwise inter-site similarities were computed, generating an inter-site similarity matrix (ISM). Inter-site texture heterogeneity metrics were computed from the ISM and compared to clinical outcomes.

Results: Of the 12 inter-site texture heterogeneity metrics evaluated, those capturing the differences in texture similarities across sites were associated with shorter overall survival (inter-site similarity entropy, similarity level cluster shade, and inter-site similarity level cluster prominence; p ≤ 0.05) and incomplete surgical resection (similarity level cluster shade, inter-site similarity level cluster prominence and inter-site cluster variance; p ≤ 0.05). Neither the total number of disease sites per patient nor the overall tumour volume per patient was associated with overall survival. Amplification of 19q12 involving cyclin E1 gene (CCNE1) predominantly occurred in patients with more heterogeneous inter-site textures.

Conclusion: Quantitative metrics non-invasively capturing spatial inter-site heterogeneity may predict outcomes in patients with HGSOC.

Key points: • Calculating inter-site texture-based heterogeneity metrics was feasible • Metrics capturing texture similarities across HGSOC sites were associated with overall survival • Heterogeneity metrics were also associated with incomplete surgical resection of HGSOC.

Keywords: Ovarian cancer; Radiogenomics; Radiomics; Survival; Texture.

Conflict of interest statement

Conflict of interest:

The authors of this manuscript declare no relationships with any companies, whose products or services may be related to the subject matter of the article.

Figures

Figure 1
Figure 1
(A) Patient with Classification of Ovarian Cancer (CLOVAR) mesenchymal subtype and an overall survival of 69 months. Texture-based results within each tumor site (i), the inter-site similarity matrix (ISM) (ii), the heterogeneity tree (iii) and the schematic of the dissimilarity of the various sites compared to the ovarian mass (iv). For instance, the diaphragmatic tumor implant (#8 in ii) has the largest dissimilarity compared to the ovarian mass. (B) Patient with CLOVAR mesenchymal subtype and an overall survival of 10 months. Texture-based results within each tumor site (i), the ISM (ii), the heterogeneity tree (iii) and the schematic of the dissimilarity of the various sites compared to the ovarian mass (iv). For instance, the left upper quadrant (LUQ) (#5 in b) has the largest dissimilarity compared to the ovarian mass, followed by the diaphragmatic and the omentum tumor implants. The numbers listed in the x axis of both figures indicates the numerical codes for lesion location (1=primary ovarian mass, 2=cul de sac, 4=omentum, 5=left upper quadrant, 7=gastro-hepatic ligament, 8=diaphragm).
Figure 1
Figure 1
(A) Patient with Classification of Ovarian Cancer (CLOVAR) mesenchymal subtype and an overall survival of 69 months. Texture-based results within each tumor site (i), the inter-site similarity matrix (ISM) (ii), the heterogeneity tree (iii) and the schematic of the dissimilarity of the various sites compared to the ovarian mass (iv). For instance, the diaphragmatic tumor implant (#8 in ii) has the largest dissimilarity compared to the ovarian mass. (B) Patient with CLOVAR mesenchymal subtype and an overall survival of 10 months. Texture-based results within each tumor site (i), the ISM (ii), the heterogeneity tree (iii) and the schematic of the dissimilarity of the various sites compared to the ovarian mass (iv). For instance, the left upper quadrant (LUQ) (#5 in b) has the largest dissimilarity compared to the ovarian mass, followed by the diaphragmatic and the omentum tumor implants. The numbers listed in the x axis of both figures indicates the numerical codes for lesion location (1=primary ovarian mass, 2=cul de sac, 4=omentum, 5=left upper quadrant, 7=gastro-hepatic ligament, 8=diaphragm).
Figure 2
Figure 2
Relationship between (A) the total number of disease sites per patient, inter-site texture heterogeneity metrics and 60 month survival and (B) the overall tumor volume per patient (calculated from CT as the sum of the volume from each segmented individual tumor site per patient), inter-site texture heterogeneity metrics and 60 month survival.
Figure 2
Figure 2
Relationship between (A) the total number of disease sites per patient, inter-site texture heterogeneity metrics and 60 month survival and (B) the overall tumor volume per patient (calculated from CT as the sum of the volume from each segmented individual tumor site per patient), inter-site texture heterogeneity metrics and 60 month survival.
Figure 3
Figure 3
Relation between Inter-site tumor Texture Heterogeneity Metrics and Genomic Alterations. (A) Heat map showing the relative similarities of the patients with respect to each other computed using the inter-site texture heterogeneity metrics. Similarity between two patients was computed using Euclidean distance measure after standardizing the features using z-score normalization. Two patients that are highly similar have a value close to 0 (red). (B) Patients were divided into two groups; the high risk group was defined as patients who underwent incomplete resection or had CCNE1 amplification and had an overall survival of ≤60 months. Conversely low risk group consisted of patients who underwent complete resection and had a survival of >60 months and no CCNE1 amplification. The SCP, SE and SCV values for each patient in the low and high risk categories according to the clinical variables and the model (classifier) are shown.
Figure 3
Figure 3
Relation between Inter-site tumor Texture Heterogeneity Metrics and Genomic Alterations. (A) Heat map showing the relative similarities of the patients with respect to each other computed using the inter-site texture heterogeneity metrics. Similarity between two patients was computed using Euclidean distance measure after standardizing the features using z-score normalization. Two patients that are highly similar have a value close to 0 (red). (B) Patients were divided into two groups; the high risk group was defined as patients who underwent incomplete resection or had CCNE1 amplification and had an overall survival of ≤60 months. Conversely low risk group consisted of patients who underwent complete resection and had a survival of >60 months and no CCNE1 amplification. The SCP, SE and SCV values for each patient in the low and high risk categories according to the clinical variables and the model (classifier) are shown.

References

    1. Engel J, Eckel R, Schubert-Fritschle G, Kerr J, Kuhn W, Diebold J, Kimmig R, Rehbock J, Holzel D. Moderate progress for ovarian cancer in the last 20 years: prolongation of survival, but no improvement in the cure rate. Eur J Cancer. 2002;38(18):2435–2445.
    1. Hyman DM, Zhou Q, Iasonos A, Grisham RN, Arnold AG, Phillips MF, Bhatia J, Levine DA, Aghajanian C, Offit K, et al. Improved survival for BRCA2-associated serous ovarian cancer compared with both BRCA-negative and BRCA1-associated serous ovarian cancer. Cancer. 2012;118(15):3703–3709.
    1. Tothill RW, Tinker AV, George J, Brown R, Fox SB, Lade S, Johnson DS, Trivett MK, Etemadmoghadam D, Locandro B, et al. Novel molecular subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clin Cancer Res. 2008;14(16):5198–5208.
    1. Verhaak RG, Tamayo P, Yang JY, Hubbard D, Zhang H, Creighton CJ, Fereday S, Lawrence M, Carter SL, Mermel CH, et al. Prognostically relevant gene signatures of high-grade serous ovarian carcinoma. The Journal of clinical investigation. 2013;123(1):517–525.
    1. Soslow RA. Histologic subtypes of ovarian carcinoma: an overview. Int J Gynecol Pathol. 2008;27(2):161–174.
    1. Khalique L, Ayhan A, Weale ME, Jacobs IJ, Ramus SJ, Gayther SA. Genetic intra-tumour heterogeneity in epithelial ovarian cancer and its implications for molecular diagnosis of tumours. J Pathol. 2007;211(3):286–295.
    1. Kumar V, Gu Y, Basu S, Berglund A, Eschrich SA, Schabath MB, Forster K, Aerts HJ, Dekker A, Fenstermacher D, et al. Radiomics: the process and the challenges. Magn Reson Imaging. 2012;30(9):1234–1248.
    1. Haralick RM, Shanmugam K, Dinstein IH. Textural Features for Image Classification. Systems, Man and Cybernetics, IEEE Transactions on. 1973;SMC-3(6):610–621.
    1. Aerts HJ, Velazquez ER, Leijenaar RT, Parmar C, Grossmann P, Carvalho S, Bussink J, Monshouwer R, Haibe-Kains B, Rietveld D, et al. Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach. Nat Commun. 2014;5:4006.
    1. Segal E, Sirlin CB, Ooi C, Adler AS, Gollub J, Chen X, Chan BK, Matcuk GR, Barry CT, Chang HY, et al. Decoding global gene expression programs in liver cancer by noninvasive imaging. Nat Biotechnol. 2007;25(6):675–680.
    1. Vargas HA, Micco M, Hong SI, Goldman DA, Dao F, Weigelt B, Soslow RA, Hricak H, Levine DA, Sala E. Association between morphologic CT imaging traits and prognostically relevant gene signatures in women with high-grade serous ovarian cancer: a hypothesis-generating study. Radiology. 2015;274(3):742–751.
    1. Veeraraghavan HSP, Papanikolopoulos NP. Adaptive templates for feature matching. Proc IEEE Intl Conf on Robotics and Automation. 2006:3393–3398.
    1. Tibshirani R. Regression shrinkage and selection via the lasso. Journal of the Royal Statistical Society Series B (Methodological) 1996:267–288.
    1. Ture MTF, Kurt I. Using Kaplan-Meier Analysis Together with Decision Tree Methods in Determining Recurrence-Free Survival of Breast Cancer Patients. Expert Systems with Applications. 2009;36:2017–2026.
    1. Etemadmoghadam D, deFazio A, Beroukhim R, Mermel C, George J, Getz G, Tothill R, Okamoto A, Raeder MB, Harnett P, et al. Integrated genome-wide DNA copy number and expression analysis identifies distinct mechanisms of primary chemoresistance in ovarian carcinomas. Clinical cancer research : an official journal of the American Association for Cancer Research. 2009;15(4):1417–1427.
    1. Patch AM, Christie EL, Etemadmoghadam D, Garsed DW, George J, Fereday S, Nones K, Cowin P, Alsop K, Bailey PJ, et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature. 2015;521(7553):489–494.
    1. Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Tarpey P, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. The New England journal of medicine. 2012;366(10):883–892.
    1. Juric D, Castel P, Griffith M, Griffith OL, Won HH, Ellis H, Ebbesen SH, Ainscough BJ, Ramu A, Iyer G, et al. Convergent loss of PTEN leads to clinical resistance to a PI(3)Kalpha inhibitor. Nature. 2015;518(7538):240–244.
    1. Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao W, Kris MG, Miller VA, Ladanyi M, Riely GJ. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res. 2013;19(8):2240–2247.
    1. Schwarz RF, Ng CK, Cooke SL, Newman S, Temple J, Piskorz AM, Gale D, Sayal K, Murtaza M, Baldwin PJ, et al. Spatial and temporal heterogeneity in high-grade serous ovarian cancer: a phylogenetic analysis. PLoS medicine. 2015;12(2):e1001789.
    1. Mittempergher L. Genomic Characterization of High-Grade Serous Ovarian Cancer: Dissecting Its Molecular Heterogeneity as a Road Towards Effective Therapeutic Strategies. Current oncology reports. 2016;18(7):44.
    1. Paracchini L, Mannarino L, Craparotta I, Romualdi C, Fruscio R, Grassi T, Fotia V, Caratti G, Perego P, Calura E, et al. Regional and temporal heterogeneity of epithelial ovarian cancer tumor biopsies: implications for therapeutic strategies. Oncotarget. 2016
    1. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science (New York, NY) 2015;348(6230):124–128.
    1. Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, Dawson N, O’Donnell PH, Balmanoukian A, Loriot Y, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet (London, England) 2016;387(10031):1909–1920.
    1. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, Berent-Maoz B, Pang J, Chmielowski B, Cherry G, et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell. 2016;165(1):35–44.

Source: PubMed

3
Abonnieren