Radiomics in Oncology

Evis Sala, Nickolas Papanikolaou

Abstract


T

umour histology classification is based on biopsy, that is invasive, destructive (reducing the number of monitoring opportunities) and suffers from poor cost efficiency. Biopsy sampling of a random spatial subregion of a tumour at a single time point may not be able to reflect the complex tumour state accurately [1-4]. Furthermore, it is well known that a hallmark of tumours is their spatial and temporal heterogeneity. On the other hand, imaging provides an opportunity to extract valuable information regarding tumour characteristics in a non-invasive way. It’s not subjected to bias selection, since the entire tumour can be assessed multiple times during the course of the disease (before, during and after treatment). However, currently imaging evaluation is based on the subjective opinion of radiologists, is time consuming, varies significantly protocol-wise and therefore suffers from low reproducibility.

Over the past decade the advances in computational image analysis methods have provided a unique opportunity to transform digital standard of care medical images to mineable high dimensional data (radiomics) that potentially reflect tumour biology and predict patient outcome in several tumour types [2, 5-15]. A relatively small number of studies have addressed the critical question of whether radiomic metrics correlate with histopathological and genomic changes in regions of interest [5, 15-18].

In this issue of the Hellenic Journal of Radiology, there are two very interesting articles on radiomics in oncology. Manikis and colleagues [19] investigate the role of T2- based MRI radiomic features for discriminating tumour grading in soft tissues sarcomas. Bisdas and colleagues [20] provide a comprehensive systematic review on the current evidence for the clinical value of radiogenomics in glioblastomas. Both papers highlight the potential important role that radiomics can play in oncology. However, existing radiomic approaches have not encoded the extent of variability between different regions within the tumour (habitats) [21, 22] and between multiple metastatic tumour sites within the patient [23]. Yet, genomic heterogeneity within the tumour and across metastatic tumour sites in the same patient is a major cause of treatment failure and development of resistance to targeted therapies [24-27] as well as specific patterns of malignant cell spread within the peritoneal cavity [28]. The lesion-specific properties, immunological components of the tumour microenvironment, may modulate malignant cell invasion and expansion, thereby shaping evolutionary selection [27,29]. However, quantification by repeated multiple tissue sampling in the same patient is challenging to implement in routine clinical practice.

Standard-of-care imaging offers a unique opportunity to non-invasively quantify and dynamically track spatial tumour heterogeneity. Nevertheless, most of the radiomics methods to date have been developed and applied to measure average intra-tumour heterogeneity based on a single disease site per patient in primary tumours [5, 15-18]. In the metastatic setting, the largest metastasis has been typically chosen for radiomics analysis and thought to be representative of the overall tumour burden heterogeneity [15, 18, 30]. In addition, most of the studies lack robust biological validation due to poor methodology for radiomics feature extraction, retrospective design and lack of adequate methods for accurate spatial co-registration of imaging with tissue sampling [11]. Most importantly, radiomics research is still working in the space of correlation rather than integration with other multi-omics data. The fact that tumours displays spatial heterogeneity at such disparate physical scales suggests that a combined approach to integrate the relevant data sources (genomics, transcriptomics, radiomics) is needed to unravel the complexity of the disease.

Full Text:

PDF

References


Wu W, Parmar C, Grossmann P, et al. Exploratory study to identify radiomics classifiers for lung cancer histology. Front Oncol 2016; 6: 71.

Aerts HJ. The potential of radiomic-based phenotyping in precision medicine: A Review. JAMA Oncol 2016; 2: 1636-1642.

Parmar C, Grossmann P, Bussink J, et al. Machine learning methods for quantitative radiomic biomarkers. Sci Rep 2015; 5: 13087.

Parmar C, Grossmann P, Rietveld D, et al. Radiomic machine-learning classifiers for prognostic biomarkers of head and neck cancer. Front Oncol 2015; 5: 272.

Segal E, Sirlin CB, Ooi C, et al. Decoding global gene expression programs in liver cancer by noninvasive imaging. Nat Biotechnol 2007; 25: 675-680.

Kumar V, Gu Y, Basu S, et al. Radiomics: the process and the challenges. Magn Reson Imaging 2012; 30: 1234-1248.

Lambin P, Rios-Velazquez E, Leijenaar R, et al. Radiomics: extracting more information from medical images using advanced feature analysis. Eur J Cancer 2012; 48: 441-446.

Aerts HJ, Velazquez ER, Leijenaar RT, et al. Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach. Nat Commun 2014; 5: 4006.

Gillies RJ, Kinahan PE, Hricak H. Radiomics: Images are more than pictures, they are data. Radiology 2016; 278: 563-577.

Lambin P, Leijenaar RTH, Deist TM, et al. Radiomics: the bridge between medical imaging and personalized medicine. Nat Rev Clin Oncol 2017; 14: 749-762.

Sanduleanu S, Woodruff HC, de Jong EEC, et al. Tracking tumor biology with radiomics: A systematic review utilizing a radiomics quality score. Radiother Oncol 2018; 127: 349-360.

Rizzo S, Botta F, Raimondi S, et al. Radiomics of high-grade serous ovarian cancer: association between quantitative CT features, residual tumour and disease progression within 12 months. Eur Radiol 2018; 28: 4849-4859.

Vargas HA, Micco M, Hong SI, et al. 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: 742-751.

Vargas HA, Veeraraghavan H, Micco M, et al. A novel representation of inter-site tumour heterogeneity from pre-treatment computed tomography textures classifies ovarian cancers by clinical outcome. Eur Radiol 2017; 27: 3991-4001.

Lu H, Arshad M, Thornton A, et al. A mathematical-descriptor of tumor-mesoscopic-structure from computed-tomography images annotates prognostic- and molecular-phenotypes of epithelial ovarian cancer. Nat Commun 2019; 10: 764.

Diehn M, Nardini C, Wang DS, et al. Identification of noninvasive imaging surrogates for brain tumor gene-expression modules. Proc Natl Acad Sci USA 2008; 105: 5213-5218.

Zhou M, Leung A, Echegaray S, et al. Non-Small Cell Lung Cancer radiogenomics map identifies relationships between molecular and imaging phenotypes with prognostic implications. Radiology 2018; 286: 307-315.

Sun R, Limkin EJ, Vakalopoulou M, et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol 2018; 19: 1180-1191.

Manikis GC, Nikiforaki K, Lagoudaki E, de Bree E, Maris TG, Marias K, Karantanas AH. T2-based MRI radiomic features for discriminating tumour grading in soft tissues sarcomas. Hell J Radiol 2019; 4(3): 11-17.

Bisdas S, Ioannidou E, D’Arco F. A Systematic review on the current radiogenomics dtudies in glioblastomas. Hell J Radiol 2019; 4(3): 32-44.

Gatenby RA, Grove O, Gillies RJ. Quantitative imaging in cancer evolution and ecology. Radiology 2013; 269: 8-15.

Cherezov D, Goldgof D, Hall L, et al. Revealing tumor habitats from texture heterogeneity analysis for classification of lung cancer malignancy and aggressiveness. Scientific Reports 2019; 9: 4500.

Sala E, Mema E, Himoto Y, et al. Unravelling tumour heterogeneity using next-generation imaging: radiomics, radiogenomics, and habitat imaging. Clin Radiol 2017; 72: 3-10.

Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 2012; 366: 883-892.

Swanton C. Intratumor heterogeneity: evolution through space and time. Cancer Res 2012; 72: 4875-4882.

Schwarz RF, Ng CK, Cooke SL, et al. Spatial and temporal heterogeneity in high-grade serous ovarian cancer: a phylogenetic analysis. PLoS Med 2015; 12: e1001789.

Zhang AW, McPherson A, Milne K, et al. Interfaces of malignant and immunologic clonal dynamics in ovarian cancer. Cell 2018; 173: 1755-1769.e1722.

Bashashati A, Ha G, Tone A, et al. Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling. J Pathol 2013; 231: 21-34.

Jimenez-Sanchez A, Memon D, Pourpe S, et al. Heterogeneous tumor-immune microenvironments among differentially growing metastases in an ovarian cancer patient. Cell 2017; 170: 927-938 e920.

Smith AD, Gray MR, del Campo SM, et al. Predicting overall survival in patients with metastatic melanoma on antiangiogenic therapy and RECIST stable disease on initial posttherapy images using CT texture analysis. AJR Am J Roentgenol 2015; 205: W283-293.




DOI: http://dx.doi.org/10.36162/hjr.v4i3.323

Refbacks

  • There are currently no refbacks.