Open Access
11 December 2015 Special Section Guest Editorial:Radiomics and Imaging Genomics: Quantitative Imaging for Precision Medicine
Author Affiliations +

Throughout the history of radiology—a medical specialty that came into being shortly after the discovery of x rays in 1895—its practice involved a skilled observer (the radiologist) looking at images and transcribing observations in relation to the indications for the imaging examination and any incidental findings. Radiologists are trained to understand how appearance on imaging correlates with underlying disease/health and strive to report it in unambiguous terms. However, there is variation in interpretation among radiologists,1,2 and even among radiologists speaking the same language, descriptive terminology varies,3,4 thereby making impractical the mass mining of radiological interpretations for discovery of linkages between observations and specific diseases.

Despite these limitations, radiologists continued to study and report on the linkage between specific image features and underlying disease, e.g., contrast enhancement patterns of focal liver lesions on CT and malignant/benign classifications of tumors on breast images. While radiologists were busy understanding and characterizing these “imaging phenotypes,” biologists were making great strides understanding the genomic basis of intracellular processes,5 leading to the ability to characterize the “molecular phenotype” (“-omics,” e.g., genomics, proteomics, metabolomics, transcriptomics, copy number, methylation) through advanced sequencing of tissue from biopsy and/or resection samples.

In the 1980s and 1990s, quantitative imaging scientists and engineers were developing algorithms for the extraction of imaging phenotypes from radiographic images for use in computer-aided detection/diagnosis and for risk assessment and prognostic/predictive tasks.6,7 However, it wasn’t until the early part of the century when researchers began exploring links between the imaging and molecular phenotypes. For example, in 2002, Huo et al. showed the relationship between computerized texture analysis of the breast parenchyma on mammography and presence of the BRAC1/BRCA2 gene mutation.8 In 2007, Segal et al. reported that radiological observations of tumors seen on CT “systematically correlate with the global gene expression programs of primary human liver cancer” derived using microarray analysis of the resected tumor.9 In 2008, Diehn et al. reported linkages between the imaging phenotype of glioblastoma multiforme (GBM) on MRI to the molecular phenotype derived using DNA microarray analysis10 and survival. And in 2010, Bhooshan et al. demonstrated relationships between computer-extracted MRI phenotypes and breast cancer subtype and aggressiveness.11 Many papers have since expanded the literature on deriving quantitative image features, deriving and reducing the interobserver variability of semantic image features, associating image features with molecular phenotypes, genetics, and outcomes, and the results of mining these associations for discovery (e.g., see Refs. 1213.14.15.16.17.18).

These and other early studies gave birth to two terms that are increasingly prevalent in the literature today. Radiomics19,20 is a name given to the science of converting medical images into computer-accessible and -searchable data. While the term radiogenomics has previously been used to describe the study of genetic variation associated with response to radiation (radiation genomics),21 in the present context we use radiogenomics (or imaging genomics) to describe relationships between molecular and imaging phenotypes.22 To highlight recent ongoing work in the areas covered by these terms, and promoted through the efforts of various programs including the National Cancer Institute’s Quantitative Imaging Network (QIN),23 the Quantitative Imaging Biomarkers Alliance (QIBA),24 and the American Association of Physicists in Medicine (AAPM),25 this issue of the Journal of Medical Imaging contains a Special Section on Radiomics and Imaging Genomics.

These ten JMI articles describe advances in radiomics and imaging genomics along several fronts. Nyflot et al. and Echegaray et al. explore variations in radiomic signatures as a function of stochastic noise and region-of-interest segmentation, respectively. Nyflot concludes that radiomics studies should specify standard acquisition protocols, while Echegaray demonstrates that there may be many radiomics features (specifically some gray-value statistics and textures) that are minimally affected by differences in segmentation boundaries.

Also within this special section, the value of one-dimensional gray-value statistics, as well as multiscale and -orientation gray-level variations (i.e., image textures), are demonstrated for several purposes. For example, Lee et al. apply these metrics to tumor habitats (regions with different intensity characteristics) in MR scans of patients with GBM, and show associations with 12-month survival. Ghosh et al. show that texture features of tumors in CT scans of patients with clear cell renal carcinoma can predict specific gene mutations. Mattonen et al. show that the image texture within automatically generated regions of interest in CT scans of patients who have had stereotactic ablative radiotherapy for lung cancer treatment can be used to separate radiation necrosis from recurrence. Tiwari et al. use texture metrics on different types of MRI scans of patients treated by laser ablation for neuropathic cancer pain that were predictive of early treatment response. Finally, while most studies of texture have been centered on the tumors themselves, Dilger et al. show that texture metrics computed from regions of interest surrounding lung nodules have value in the prediction of malignancy.

Other investigators report novel frameworks for integrating radiomic and -omics data and mining the resulting databases for associations with clinical data. For example, for breast cancer, Wu et al. integrate mammographic features and SNPs with traditional risk factors to improve risk prediction, and Guo et al. show significant correlations of DCE-MRI radiomic features to clinical and genomic characteristics. Both of these and many other studies argue for continued development and expansion of large imaging26 and -omics27 databases utilizing standardized protocols. Finally, lest one conclude that image features are only useful in cancer research, see Xie et al. for a report on detecting ventricular-septal defects in mouse embryos through segmentation and pixel analysis.

A word of caution, however. While radiomics and imaging genomics articles continue to populate the literature, many of them (including some in this special section) (a) involve small numbers of subjects with respect to the number of radiomics features investigated, thereby raising concerns of over fitting; or (b) do not report validations in external cohorts, thereby limiting generalizability to additional patient populations, imaging by different scanner types, etc. These articles are important landmarks and vehicles for disseminating ideas, but themselves should be seen as pilot studies, suggestive of further investigation and validation. Those of us in this research community should remain conscious that correlation does not imply causation28 and that we need to strive to fully validate and generalize our methods and results.

References

1. 

and III S. G. Armato et al., “The Lung Image Database Consortium (LIDC): an evaluation of radiologist variability in the identification of lung nodules on CT scans,” Acad. Radiol., 14 (11), 1409 –1421 (2007). http://dx.doi.org/10.1016/j.acra.2007.07.008 Google Scholar

2. 

B. J. Hillman et al., “Improving diagnostic accuracy: a comparison of interactive and Delphi consultations,” Invest. Radiol., 12 (2), 112 –115 (1977). http://dx.doi.org/10.1097/00004424-197703000-00002 INVRAV 0020-9996 Google Scholar

3. 

H. J. Lowe et al., “Automated semantic indexing of imaging reports to support retrieval of medical images in the multimedia electronic medical record,” Methods Inf. Med., 38 (4–5), 303 –307 (1999). Google Scholar

4. 

D. Korenblum et al., “Managing biomedical image metadata for search and retrieval of similar images,” J. Digit. Imaging, 24 (4), 739 –748 (2011). http://dx.doi.org/10.1007/s10278-010-9328-z Google Scholar

5. 

R. Mirnezami, J. Nicholson and A. Darzi, “Preparing for precision medicine,” N. Engl. J. Med., 366 (6), 489 –491 (2012). http://dx.doi.org/10.1056/NEJMp1114866 NEJMAG 0028-4793 Google Scholar

6. 

M. L. Giger, H.P. Chan and J. Boone, “Anniversary paper: history and status of CAD and quantitative image analysis: the role of medical physics and AAPM,” Med. Phys., 35 (12), 5799 –5820 (2008). http://dx.doi.org/10.1118/1.3013555 MPHYA6 0094-2405 Google Scholar

7. 

M. L. Giger, N. Karssemeijer and J. A. Schnabel, “Breast image analysis for risk assessment, detection, diagnosis, and treatment of cancer,” Annu. Rev. Biomed. Eng., 15 327 –357 (2013). http://dx.doi.org/10.1146/annurev-bioeng-071812-152416 ARBEF7 1523-9829 Google Scholar

8. 

Z. Huo et al., “Computerized analysis of digitized mammograms of BRCA1 and BRCA2 gene mutation carriers,” Radiology, 225 (2), 519 –526 (2002). http://dx.doi.org/10.1148/radiol.2252010845 RADLAX 0033-8419 Google Scholar

9. 

E. Segal et al., “Decoding global gene expression programs in liver cancer by noninvasive imaging,” Nat. Biotechnol., 25 675 –680 (2007). http://dx.doi.org/10.1038/nbt1306 NABIF9 1087-0156 Google Scholar

10. 

M. Diehn et al., “Identification of noninvasive imaging surrogates for brain tumor gene-expression modules,” Proc. Nat. Acad. Sci. U.S.A., 105 (13), 5213 –5218 (2008). http://dx.doi.org/10.1073/pnas.0801279105 PNASA6 0027-8424 Google Scholar

11. 

N. Bhooshan et al., “Cancerous breast lesions on dynamic contrast-enhanced MR images: computerized characterization for image-based prognostic markers,” Radiology, 254 (3), 680 –690 (2010). http://dx.doi.org/10.1148/radiol.09090838 RADLAX 0033-8419 Google Scholar

12. 

O. Gevaert et al., “Non-small cell lung cancer: identifying prognostic imaging biomarkers by leveraging public gene expression microarray data—methods and preliminary results,” Radiology, 264 (2), 387 –396 (2012). http://dx.doi.org/10.1148/radiol.12111607 RADLAX 0033-8419 Google Scholar

13. 

H. J. Aerts et al., “Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach,” Nat. Commun., 5 4006 (2014). http://dx.doi.org/10.1038/ncomms5006 Google Scholar

14. 

R. Colen et al., “NCI workshop report: clinical and computational requirements for correlating imaging phenotypes with genomics signatures,” Transl. Oncol., 7 (5), 556 –569 (2014). http://dx.doi.org/10.1016/j.tranon.2014.07.007 Google Scholar

15. 

H. Itakura et al., “Magnetic resonance image features identify glioblastoma phenotypic subtypes with distinct molecular pathway activities,” Sci. Transl. Med., 7 (303), 303ra138 (2015). http://dx.doi.org/10.1126/scitranslmed.aaa7582 Google Scholar

16. 

O. Grove et al., “Quantitative computed tomographic descriptors associate tumor shape complexity and intratumor heterogeneity with prognosis in lung adenocarcinoma,” PLoS One, 10 (3), e0118261 (2015). http://dx.doi.org/10.1371/journal.pone.0118261 1932-6203 Google Scholar

17. 

H. Li et al., “Pilot study demonstrating potential association between breast cancer image-based risk phenotypes and genomic biomarkers,” Med. Phys., 41 (3), 031917 (2014). http://dx.doi.org/10.1118/1.4865811 MPHYA6 0094-2405 Google Scholar

18. 

C. C. Jaffe, “Imaging and genomics: is there a synergy?,” Radiology, 264 (2), 329 –331 (2012). http://dx.doi.org/10.1148/radiol.12120871 RADLAX 0033-8419 Google Scholar

19. 

P. Lambin et al., “Extracting more information from medical images using advanced feature analysis,” Eur. J. Cancer, 48 (4), 441 –446 (2012). http://dx.doi.org/10.1016/j.ejca.2011.11.036 EJCAEL 0959-8049 Google Scholar

20. 

V. Kumar et al., “Radiomics: the process and the challenges,” Magn. Reson. Imaging, 30 (9), 1234 –1248 (2012). http://dx.doi.org/10.1016/j.mri.2012.06.010 MRIMDQ 0730-725X Google Scholar

21. 

N. G. Burnet et al., “Radiosensitivity, radiogenomics and RAPPER,” Clin. Oncol., 18 (7), 525 –528 (2006). http://dx.doi.org/0.1016/j.clon.2006.05.007 Google Scholar

22. 

A. M. Rutman and M. D. Kuo, “Radiogenomics: creating a link between molecular diagnostics and diagnostic imaging,” Eur. J. Radiol., 70 (2), 232 –241 (2009). http://dx.doi.org/10.1016/j.ejrad.2009.01.050 EJRADR 0720-048X Google Scholar

23. 

L. P. Clarke et al., “The quantitative imaging network: NCI’s historical perspective and planned goals,” Transl. Oncol., 7 1 –4 (2014). http://dx.doi.org/10.1593/tlo.13832 Google Scholar

24. 

A. J. Buckler et al., “Quantitative imaging test approval and biomarker qualification: interrelated but distinct activities,” Radiology, 259 (3), 875 –884 (2011). http://dx.doi.org/10.1148/radiol.10100800 RADLAX 0033-8419 Google Scholar

25. 

AAPM FOREM on Imaging Genomics, Conference Agenda, 30 September–1 October 2014, Houston, Texas (2015) http://www.aapm.org/meetings/documents/revfinalAgendaforFOREM09242014.pdf November ). 2015). Google Scholar

26. 

K. Clark et al., “The Cancer Imaging Archive (TCIA): maintaining and operating a public information repository,” J. Digit. Imaging, 26 (6), 1045 –1057 (2013). http://dx.doi.org/10.1007/s10278-013-9622-7 Google Scholar

27. 

K. Tomczak, P. Czerwinska and M. Wiznerowicz, “The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge,” Contemp Oncol., 19 (1A), A68 –77 (2015). http://dx.doi.org/10.5114/wo.2014.47136 Google Scholar

28. 

M. D. Kuo and N. Jamshidi, “Behind the numbers: decoding molecular phenotypes with radiogenomics—guiding principles and technical considerations,” Radiology, 270 (2), 320 –325 (2014). http://dx.doi.org/10.1148/radiol.13132195 RADLAX 0033-8419 Google Scholar

Biography

Sandy Napel received his BSES from SUNY Stony Brook in 1974 and his MSEE and PhD in EE from Stanford University in 1976 and 1981, respectively. He was formerly VP of engineering at Imatron Inc, and is currently a professor of radiology and, by courtesy, of electrical engineering and medicine (biomedical informatics research) at Stanford University. He co-leads the Stanford Radiology 3D and Quantitative Imaging Lab and the Radiology Department’s Section on Integrative Biomedical Imaging Informatics, where he is developing techniques for linkage of image features to molecular properties of disease.

Maryellen Giger received her BS from Illinois Benedictine College in 1978; MSc from University of Exeter, England, in 1979; and PhD from University of Chicago in 1985. She is the A. N. Pritzker Professor of Radiology of the Committee on Medical Physics and the College at The University of Chicago. She is vice chair of radiology (basic science research) and leads an NIH-funded lab on computer-aided diagnosis and radiomics in collaboration with other scientists to develop predictive image-based signatures for precision medicine.

© 2015 Society of Photo-Optical Instrumentation Engineers (SPIE)
Sandy Napel and Maryellen L. Giger "Special Section Guest Editorial:Radiomics and Imaging Genomics: Quantitative Imaging for Precision Medicine," Journal of Medical Imaging 2(4), 041001 (11 December 2015). https://doi.org/10.1117/1.JMI.2.4.041001
Published: 11 December 2015
Lens.org Logo
CITATIONS
Cited by 17 scholarly publications.
Advertisement
Advertisement
RIGHTS & PERMISSIONS
Get copyright permission  Get copyright permission on Copyright Marketplace
KEYWORDS
Medicine

Tumors

Magnetic resonance imaging

Cancer

Image segmentation

Mining

Radiology

Back to Top