301
|
Grossmann P, Stringfield O, El-Hachem N, Bui MM, Rios Velazquez E, Parmar C, Leijenaar RTH, Haibe-Kains B, Lambin P, Gillies RJ, Aerts HJWL. Defining the biological basis of radiomic phenotypes in lung cancer. eLife 2017; 6:e23421. [PMID: 28731408 PMCID: PMC5590809 DOI: 10.7554/elife.23421] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 07/17/2017] [Indexed: 02/06/2023] Open
Abstract
Medical imaging can visualize characteristics of human cancer noninvasively. Radiomics is an emerging field that translates these medical images into quantitative data to enable phenotypic profiling of tumors. While radiomics has been associated with several clinical endpoints, the complex relationships of radiomics, clinical factors, and tumor biology are largely unknown. To this end, we analyzed two independent cohorts of respectively 262 North American and 89 European patients with lung cancer, and consistently identified previously undescribed associations between radiomic imaging features, molecular pathways, and clinical factors. In particular, we found a relationship between imaging features, immune response, inflammation, and survival, which was further validated by immunohistochemical staining. Moreover, a number of imaging features showed predictive value for specific pathways; for example, intra-tumor heterogeneity features predicted activity of RNA polymerase transcription (AUC = 0.62, p=0.03) and intensity dispersion was predictive of the autodegration pathway of a ubiquitin ligase (AUC = 0.69, p<10-4). Finally, we observed that prognostic biomarkers performed highest when combining radiomic, genetic, and clinical information (CI = 0.73, p<10-9) indicating complementary value of these data. In conclusion, we demonstrate that radiomic approaches permit noninvasive assessment of both molecular and clinical characteristics of tumors, and therefore have the potential to advance clinical decision-making by systematically analyzing standard-of-care medical images.
Collapse
Affiliation(s)
- Patrick Grossmann
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, United States
| | - Olya Stringfield
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States
| | - Nehme El-Hachem
- Integrative systems biology, Institut de recherches cliniques de Montreal, Montreal, Canada.
| | - Marilyn M Bui
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States
| | - Emmanuel Rios Velazquez
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| | - Chintan Parmar
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of Radiation Oncology, Research Institute GROW, Maastricht University, Maastricht, Netherlands
| | - Ralph TH Leijenaar
- Department of Radiation Oncology, Research Institute GROW, Maastricht University, Maastricht, Netherlands
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
- Medical Biophysics Department, University of Toronto, Toronto, Canada
| | - Philippe Lambin
- Department of Radiation Oncology, Research Institute GROW, Maastricht University, Maastricht, Netherlands
| | - Robert J Gillies
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States
| | - Hugo JWL Aerts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, United States
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
302
|
Wu J, Cui Y, Sun X, Cao G, Li B, Ikeda DM, Kurian AW, Li R. Unsupervised Clustering of Quantitative Image Phenotypes Reveals Breast Cancer Subtypes with Distinct Prognoses and Molecular Pathways. Clin Cancer Res 2017; 23:3334-3342. [PMID: 28073839 PMCID: PMC5496801 DOI: 10.1158/1078-0432.ccr-16-2415] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/29/2016] [Accepted: 01/03/2017] [Indexed: 01/28/2023]
Abstract
Purpose: To identify novel breast cancer subtypes by extracting quantitative imaging phenotypes of the tumor and surrounding parenchyma and to elucidate the underlying biologic underpinnings and evaluate the prognostic capacity for predicting recurrence-free survival (RFS).Experimental Design: We retrospectively analyzed dynamic contrast-enhanced MRI data of patients from a single-center discovery cohort (n = 60) and an independent multicenter validation cohort (n = 96). Quantitative image features were extracted to characterize tumor morphology, intratumor heterogeneity of contrast agent wash-in/wash-out patterns, and tumor-surrounding parenchyma enhancement. On the basis of these image features, we used unsupervised consensus clustering to identify robust imaging subtypes and evaluated their clinical and biologic relevance. We built a gene expression-based classifier of imaging subtypes and tested their prognostic significance in five additional cohorts with publically available gene expression data but without imaging data (n = 1,160).Results: Three distinct imaging subtypes, that is, homogeneous intratumoral enhancing, minimal parenchymal enhancing, and prominent parenchymal enhancing, were identified and validated. In the discovery cohort, imaging subtypes stratified patients with significantly different 5-year RFS rates of 79.6%, 65.2%, 52.5% (log-rank P = 0.025) and remained as an independent predictor after adjusting for clinicopathologic factors (HR, 2.79; P = 0.016). The prognostic value of imaging subtypes was further validated in five independent gene expression cohorts, with average 5-year RFS rates of 88.1%, 74.0%, 59.5% (log-rank P from <0.0001 to 0.008). Each imaging subtype was associated with specific dysregulated molecular pathways that can be therapeutically targeted.Conclusions: Imaging subtypes provide complimentary value to established histopathologic or molecular subtypes and may help stratify patients with breast cancer. Clin Cancer Res; 23(13); 3334-42. ©2017 AACR.
Collapse
Affiliation(s)
- Jia Wu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Yi Cui
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
- Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Proton Beam Therapy Center, Sapporo, Hokkaido, Japan
| | - Xiaoli Sun
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
- Radiotherapy Department, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Guohong Cao
- Department of Radiology, International Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Bailiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Debra M Ikeda
- Department of Radiology, Stanford University School of Medicine, Advanced Medicine Center, Stanford, California
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Allison W Kurian
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
303
|
Wang Q, Zhou S, Court LE, Verma V, Koay EJ, Zhang L, Zhang W, Tang C, Lin S, Welsh JD, Blum M, Betancourt S, Maru D, Hofstetter WL, Chang JY. Radiomics predicts clinical outcome in primary gastroesophageal junction adenocarcinoma treated by chemo/radiotherapy and surgery. Phys Imaging Radiat Oncol 2017. [DOI: 10.1016/j.phro.2017.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
304
|
Evolution of lymphoma staging and response evaluation: current limitations and future directions. Nat Rev Clin Oncol 2017; 14:631-645. [DOI: 10.1038/nrclinonc.2017.78] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
305
|
CT-based radiomics signature: a potential biomarker for preoperative prediction of early recurrence in hepatocellular carcinoma. Abdom Radiol (NY) 2017; 42:1695-1704. [PMID: 28180924 DOI: 10.1007/s00261-017-1072-0] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE To develop a CT-based radiomics signature and assess its ability for preoperatively predicting the early recurrence (≤1 year) of hepatocellular carcinoma (HCC). METHODS A total of 215 HCC patients who underwent partial hepatectomy were enrolled in this retrospective study, and all the patients were followed up at least within 1 year. Radiomics features were extracted from arterial- and portal venous-phase CT images, and a radiomics signature was built by the least absolute shrinkage and selection operator (LASSO) logistic regression model. Preoperative clinical factors associated with early recurrence were evaluated. A radiomics signature, a clinical model, and a combined model were built, and the area under the curve (AUC) of operating characteristics (ROC) was used to explore their performance to discriminate early recurrence. RESULTS Twenty-one radiomics features were chosen from 300 candidate features to build a radiomics signature that was significantly associated with early recurrence (P < 0.001), and they presented good performance in the discrimination of early recurrence alone with an AUC of 0.817 (95% CI: 0.758-0.866), sensitivity of 0.794, and specificity of 0.699. The AUCs of the clinical and combined models were 0.781 (95% CI: 0.719-0.834) and 0.836 (95% CI: 0.779-0.883), respectively, with the sensitivity being 0.784 and 0.824, and the specificity being 0.619 and 0.708, respectively. Adding a radiomics signature into conventional clinical variables can significantly improve the accuracy of the preoperative model in predicting early recurrence (P = 0.01). CONCLUSIONS The radiomics signature was a significant predictor for early recurrence in HCC. Incorporating radiomics signature into conventional clinical factors performed better for preoperative estimation of early recurrence than with clinical variables alone.
Collapse
|
306
|
Quantification of hepatocellular carcinoma heterogeneity with multiparametric magnetic resonance imaging. Sci Rep 2017; 7:2452. [PMID: 28550313 PMCID: PMC5446396 DOI: 10.1038/s41598-017-02706-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022] Open
Abstract
Tumour heterogeneity poses a significant challenge for treatment stratification. The goals of this study were to quantify heterogeneity in hepatocellular carcinoma (HCC) using multiparametric magnetic resonance imaging (mpMRI), and to report preliminary data correlating quantitative MRI parameters with advanced histopathology and gene expression in a patient subset. Thirty-two HCC patients with 39 HCC lesions underwent mpMRI including diffusion-weighted imaging (DWI), blood-oxygenation-level-dependent (BOLD), tissue-oxygenation-level-dependent (TOLD) and dynamic contrast-enhanced (DCE)-MRI. Histogram characteristics [central tendency (mean, median) and heterogeneity (standard deviation, kurtosis, skewness) MRI parameters] in HCC and liver parenchyma were compared using Wilcoxon signed-rank tests. Histogram data was correlated between MRI methods in all patients and with histopathology and gene expression in 14 patients. HCCs exhibited significantly higher intra-tissue heterogeneity vs. liver with all MRI methods (P < 0.030). Although central tendency parameters showed significant correlations between MRI methods and with each of histopathology and gene expression, heterogeneity parameters exhibited additional complementary correlations between BOLD and DCE-MRI and with histopathologic hypoxia marker HIF1α and gene expression of Wnt target GLUL, pharmacological target FGFR4, stemness markers EPCAM and KRT19 and immune checkpoint PDCD1. Histogram analysis combining central tendency and heterogeneity mpMRI features is promising for non-invasive HCC characterization on the imaging, histologic and genomics levels.
Collapse
|
307
|
Booth TC, Larkin TJ, Yuan Y, Kettunen MI, Dawson SN, Scoffings D, Canuto HC, Vowler SL, Kirschenlohr H, Hobson MP, Markowetz F, Jefferies S, Brindle KM. Analysis of heterogeneity in T2-weighted MR images can differentiate pseudoprogression from progression in glioblastoma. PLoS One 2017; 12:e0176528. [PMID: 28520730 PMCID: PMC5435159 DOI: 10.1371/journal.pone.0176528] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 04/12/2017] [Indexed: 01/22/2023] Open
Abstract
PURPOSE To develop an image analysis technique that distinguishes pseudoprogression from true progression by analyzing tumour heterogeneity in T2-weighted images using topological descriptors of image heterogeneity called Minkowski functionals (MFs). METHODS Using a retrospective patient cohort (n = 50), and blinded to treatment response outcome, unsupervised feature estimation was performed to investigate MFs for the presence of outliers, potential confounders, and sensitivity to treatment response. The progression and pseudoprogression groups were then unblinded and supervised feature selection was performed using MFs, size and signal intensity features. A support vector machine model was obtained and evaluated using a prospective test cohort. RESULTS The model gave a classification accuracy, using a combination of MFs and size features, of more than 85% in both retrospective and prospective datasets. A different feature selection method (Random Forest) and classifier (Lasso) gave the same results. Although not apparent to the reporting radiologist, the T2-weighted hyperintensity phenotype of those patients with progression was heterogeneous, large and frond-like when compared to those with pseudoprogression. CONCLUSION Analysis of heterogeneity, in T2-weighted MR images, which are acquired routinely in the clinic, has the potential to detect an earlier treatment response allowing an early change in treatment strategy. Prospective validation of this technique in larger datasets is required.
Collapse
Affiliation(s)
- Thomas C. Booth
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Timothy J. Larkin
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Yinyin Yuan
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Mikko I. Kettunen
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Sarah N. Dawson
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Daniel Scoffings
- Department of Radiology, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Holly C. Canuto
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Sarah L. Vowler
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Heide Kirschenlohr
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Michael P. Hobson
- Battock Centre for Experimental Astrophysics, Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Florian Markowetz
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Sarah Jefferies
- Department of Oncology, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Kevin M. Brindle
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| |
Collapse
|
308
|
Cox VL, Bhosale P, Varadhachary GR, Wagner-Bartak N, Glitza IC, Gold KA, Atkins JT, Soliman PT, Hong DS, Qayyum A. Cancer Genomics and Important Oncologic Mutations: A Contemporary Guide for Body Imagers. Radiology 2017; 283:314-340. [DOI: 10.1148/radiol.2017152224] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Veronica L. Cox
- From the Department of Radiology, Abdominal Imaging Section (V.L.C., P.B., N.W.B., A.Q.), Department of Gastrointestinal Medical Oncology (G.R.V.), Department of Melanoma Medical Oncology (I.C.G.), Department of Thoracic and Head & Neck Medical Oncology (K.A.G.), Department of Gynecologic Oncology (P.T.S.), Department of Investigational Cancer Therapeutics (J.T.A., D.S.H.), University of Texas MD
| | - Priya Bhosale
- From the Department of Radiology, Abdominal Imaging Section (V.L.C., P.B., N.W.B., A.Q.), Department of Gastrointestinal Medical Oncology (G.R.V.), Department of Melanoma Medical Oncology (I.C.G.), Department of Thoracic and Head & Neck Medical Oncology (K.A.G.), Department of Gynecologic Oncology (P.T.S.), Department of Investigational Cancer Therapeutics (J.T.A., D.S.H.), University of Texas MD
| | - Gauri R. Varadhachary
- From the Department of Radiology, Abdominal Imaging Section (V.L.C., P.B., N.W.B., A.Q.), Department of Gastrointestinal Medical Oncology (G.R.V.), Department of Melanoma Medical Oncology (I.C.G.), Department of Thoracic and Head & Neck Medical Oncology (K.A.G.), Department of Gynecologic Oncology (P.T.S.), Department of Investigational Cancer Therapeutics (J.T.A., D.S.H.), University of Texas MD
| | - Nicolaus Wagner-Bartak
- From the Department of Radiology, Abdominal Imaging Section (V.L.C., P.B., N.W.B., A.Q.), Department of Gastrointestinal Medical Oncology (G.R.V.), Department of Melanoma Medical Oncology (I.C.G.), Department of Thoracic and Head & Neck Medical Oncology (K.A.G.), Department of Gynecologic Oncology (P.T.S.), Department of Investigational Cancer Therapeutics (J.T.A., D.S.H.), University of Texas MD
| | - Isabella C. Glitza
- From the Department of Radiology, Abdominal Imaging Section (V.L.C., P.B., N.W.B., A.Q.), Department of Gastrointestinal Medical Oncology (G.R.V.), Department of Melanoma Medical Oncology (I.C.G.), Department of Thoracic and Head & Neck Medical Oncology (K.A.G.), Department of Gynecologic Oncology (P.T.S.), Department of Investigational Cancer Therapeutics (J.T.A., D.S.H.), University of Texas MD
| | - Kathryn A. Gold
- From the Department of Radiology, Abdominal Imaging Section (V.L.C., P.B., N.W.B., A.Q.), Department of Gastrointestinal Medical Oncology (G.R.V.), Department of Melanoma Medical Oncology (I.C.G.), Department of Thoracic and Head & Neck Medical Oncology (K.A.G.), Department of Gynecologic Oncology (P.T.S.), Department of Investigational Cancer Therapeutics (J.T.A., D.S.H.), University of Texas MD
| | - Johnique T. Atkins
- From the Department of Radiology, Abdominal Imaging Section (V.L.C., P.B., N.W.B., A.Q.), Department of Gastrointestinal Medical Oncology (G.R.V.), Department of Melanoma Medical Oncology (I.C.G.), Department of Thoracic and Head & Neck Medical Oncology (K.A.G.), Department of Gynecologic Oncology (P.T.S.), Department of Investigational Cancer Therapeutics (J.T.A., D.S.H.), University of Texas MD
| | - Pamela T. Soliman
- From the Department of Radiology, Abdominal Imaging Section (V.L.C., P.B., N.W.B., A.Q.), Department of Gastrointestinal Medical Oncology (G.R.V.), Department of Melanoma Medical Oncology (I.C.G.), Department of Thoracic and Head & Neck Medical Oncology (K.A.G.), Department of Gynecologic Oncology (P.T.S.), Department of Investigational Cancer Therapeutics (J.T.A., D.S.H.), University of Texas MD
| | - David S. Hong
- From the Department of Radiology, Abdominal Imaging Section (V.L.C., P.B., N.W.B., A.Q.), Department of Gastrointestinal Medical Oncology (G.R.V.), Department of Melanoma Medical Oncology (I.C.G.), Department of Thoracic and Head & Neck Medical Oncology (K.A.G.), Department of Gynecologic Oncology (P.T.S.), Department of Investigational Cancer Therapeutics (J.T.A., D.S.H.), University of Texas MD
| | - Aliya Qayyum
- From the Department of Radiology, Abdominal Imaging Section (V.L.C., P.B., N.W.B., A.Q.), Department of Gastrointestinal Medical Oncology (G.R.V.), Department of Melanoma Medical Oncology (I.C.G.), Department of Thoracic and Head & Neck Medical Oncology (K.A.G.), Department of Gynecologic Oncology (P.T.S.), Department of Investigational Cancer Therapeutics (J.T.A., D.S.H.), University of Texas MD
| |
Collapse
|
309
|
Jamshidi N, Margolis DJ, Raman S, Huang J, Reiter RE, Kuo MD. Multiregional Radiogenomic Assessment of Prostate Microenvironments with Multiparametric MR Imaging and DNA Whole-Exome Sequencing of Prostate Glands with Adenocarcinoma. Radiology 2017; 284:109-119. [PMID: 28453432 DOI: 10.1148/radiol.2017162827] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Purpose To assess the underlying genomic variation of prostate gland microenvironments of patients with prostate adenocarcinoma in the context of colocalized multiparametric magnetic resonance (MR) imaging and histopathologic assessment of normal and abnormal regions by using whole-exome sequencing. Materials and Methods Six patients with prostate adenocarcinoma who underwent robotic prostatectomy with whole-mount preservation of the prostate were identified, which enabled spatial mapping between preoperative multiparametric MR imaging and the gland. Four regions of interest were identified within each gland, including regions found to be normal and abnormal via histopathologic analysis. Whole-exome DNA sequencing (>50 times coverage) was performed on each of these spatially targeted regions. Radiogenomic analysis of imaging and mutation data were performed with hierarchical clustering, phylogenetic analysis, and principal component analysis. Results Radiogenomic multiparametric MR imaging and whole-exome spatial characterization in six patients with prostate adenocarcinoma (three patients, Gleason score of 3 + 4; and three patients, Gleason score of 4 + 5) was performed across 23 spatially distinct regions. Hierarchical clustering separated histopathologic analysis-proven high-grade lesions from the normal regions, and this reflected concordance between multiparametric MR imaging and resultant histopathologic analysis in all patients. Seventy-seven mutations involving 29 cancer-associated genes across the 23 spatially distinct prostate samples were identified. There was no significant difference in mutation load in cancer-associated genes between regions that were proven to be normal via histopathologic analysis (34 mutations per sample ± 19), mildly suspicious via multiparametric MR imaging (37 mutations per sample ± 21), intermediately suspicious via multiparametric MR imaging (31 mutations per sample ± 15), and high-grade cancer (33 mutations per sample ± 18) (P = .30). Principal component analysis resolved samples from different patients and further classified samples (regardless of histopathologic status) from prostate glands with Gleason score 3 + 4 versus 4 + 5 samples. Conclusion Multiregion spatial multiparametric MR imaging and whole-exome radiogenomic analysis of prostate glands with adenocarcinoma shows a continuum of mutations across regions that were found via histologic analysis to be high grade and normal. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Neema Jamshidi
- From the Departments of Radiological Sciences (N.J., S.R., M.D.K.) and Urology (R.E.R.), University of California, Los Angeles-David Geffen School of Medicine, 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721; Department of Radiology, Weill Cornell Imaging, New York-Presbyterian Hospital, New York, NY (D.J.M.); Department of Pathology, Duke University School of Medicine, Durham, NC (J.H.); and College of Electrical and Computer Engineering, National Chiao Tung University, HsinChu, Taiwan (M.D.K.)
| | - Daniel J Margolis
- From the Departments of Radiological Sciences (N.J., S.R., M.D.K.) and Urology (R.E.R.), University of California, Los Angeles-David Geffen School of Medicine, 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721; Department of Radiology, Weill Cornell Imaging, New York-Presbyterian Hospital, New York, NY (D.J.M.); Department of Pathology, Duke University School of Medicine, Durham, NC (J.H.); and College of Electrical and Computer Engineering, National Chiao Tung University, HsinChu, Taiwan (M.D.K.)
| | - Steven Raman
- From the Departments of Radiological Sciences (N.J., S.R., M.D.K.) and Urology (R.E.R.), University of California, Los Angeles-David Geffen School of Medicine, 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721; Department of Radiology, Weill Cornell Imaging, New York-Presbyterian Hospital, New York, NY (D.J.M.); Department of Pathology, Duke University School of Medicine, Durham, NC (J.H.); and College of Electrical and Computer Engineering, National Chiao Tung University, HsinChu, Taiwan (M.D.K.)
| | - Jiaoti Huang
- From the Departments of Radiological Sciences (N.J., S.R., M.D.K.) and Urology (R.E.R.), University of California, Los Angeles-David Geffen School of Medicine, 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721; Department of Radiology, Weill Cornell Imaging, New York-Presbyterian Hospital, New York, NY (D.J.M.); Department of Pathology, Duke University School of Medicine, Durham, NC (J.H.); and College of Electrical and Computer Engineering, National Chiao Tung University, HsinChu, Taiwan (M.D.K.)
| | - Robert E Reiter
- From the Departments of Radiological Sciences (N.J., S.R., M.D.K.) and Urology (R.E.R.), University of California, Los Angeles-David Geffen School of Medicine, 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721; Department of Radiology, Weill Cornell Imaging, New York-Presbyterian Hospital, New York, NY (D.J.M.); Department of Pathology, Duke University School of Medicine, Durham, NC (J.H.); and College of Electrical and Computer Engineering, National Chiao Tung University, HsinChu, Taiwan (M.D.K.)
| | - Michael D Kuo
- From the Departments of Radiological Sciences (N.J., S.R., M.D.K.) and Urology (R.E.R.), University of California, Los Angeles-David Geffen School of Medicine, 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721; Department of Radiology, Weill Cornell Imaging, New York-Presbyterian Hospital, New York, NY (D.J.M.); Department of Pathology, Duke University School of Medicine, Durham, NC (J.H.); and College of Electrical and Computer Engineering, National Chiao Tung University, HsinChu, Taiwan (M.D.K.)
| |
Collapse
|
310
|
Incoronato M, Aiello M, Infante T, Cavaliere C, Grimaldi AM, Mirabelli P, Monti S, Salvatore M. Radiogenomic Analysis of Oncological Data: A Technical Survey. Int J Mol Sci 2017; 18:ijms18040805. [PMID: 28417933 PMCID: PMC5412389 DOI: 10.3390/ijms18040805] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 12/18/2022] Open
Abstract
In the last few years, biomedical research has been boosted by the technological development of analytical instrumentation generating a large volume of data. Such information has increased in complexity from basic (i.e., blood samples) to extensive sets encompassing many aspects of a subject phenotype, and now rapidly extending into genetic and, more recently, radiomic information. Radiogenomics integrates both aspects, investigating the relationship between imaging features and gene expression. From a methodological point of view, radiogenomics takes advantage of non-conventional data analysis techniques that reveal meaningful information for decision-support in cancer diagnosis and treatment. This survey is aimed to review the state-of-the-art techniques employed in radiomics and genomics with special focus on analysis methods based on molecular and multimodal probes. The impact of single and combined techniques will be discussed in light of their suitability in correlation and predictive studies of specific oncologic diseases.
Collapse
Affiliation(s)
| | - Marco Aiello
- IRCCS SDN, Via E. Gianturco, 113, 80143 Naples, Italy.
| | | | | | | | | | - Serena Monti
- IRCCS SDN, Via E. Gianturco, 113, 80143 Naples, Italy.
| | | |
Collapse
|
311
|
Abstract
There is interest in identifying and quantifying tumor heterogeneity at the genomic, tissue pathology and clinical imaging scales, as this may help better understand tumor biology and may yield useful biomarkers for guiding therapy-based decision making. This review focuses on the role and value of using x-ray, CT, MRI and PET based imaging methods that identify, measure and map tumor heterogeneity. In particular we highlight the potential value of these techniques and the key challenges required to validate and qualify these biomarkers for clinical use.
Collapse
Affiliation(s)
- James P B O'Connor
- Institute of Cancer Sciences, University of Manchester, Manchester, UK; Department of Radiology, The Christie Hospital NHS Trust, Manchester, UK.
| |
Collapse
|
312
|
Lee G, Lee HY, Ko ES, Jeong WK. Radiomics and imaging genomics in precision medicine. PRECISION AND FUTURE MEDICINE 2017. [DOI: 10.23838/pfm.2017.00101] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
313
|
Gevaert O, Echegaray S, Khuong A, Hoang CD, Shrager JB, Jensen KC, Berry GJ, Guo HH, Lau C, Plevritis SK, Rubin DL, Napel S, Leung AN. Predictive radiogenomics modeling of EGFR mutation status in lung cancer. Sci Rep 2017; 7:41674. [PMID: 28139704 PMCID: PMC5282551 DOI: 10.1038/srep41674] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 12/21/2016] [Indexed: 11/18/2022] Open
Abstract
Molecular analysis of the mutation status for EGFR and KRAS are now routine in the management of non-small cell lung cancer. Radiogenomics, the linking of medical images with the genomic properties of human tumors, provides exciting opportunities for non-invasive diagnostics and prognostics. We investigated whether EGFR and KRAS mutation status can be predicted using imaging data. To accomplish this, we studied 186 cases of NSCLC with preoperative thin-slice CT scans. A thoracic radiologist annotated 89 semantic image features of each patient’s tumor. Next, we built a decision tree to predict the presence of EGFR and KRAS mutations. We found a statistically significant model for predicting EGFR but not for KRAS mutations. The test set area under the ROC curve for predicting EGFR mutation status was 0.89. The final decision tree used four variables: emphysema, airway abnormality, the percentage of ground glass component and the type of tumor margin. The presence of either of the first two features predicts a wild type status for EGFR while the presence of any ground glass component indicates EGFR mutations. These results show the potential of quantitative imaging to predict molecular properties in a non-invasive manner, as CT imaging is more readily available than biopsies.
Collapse
Affiliation(s)
- Olivier Gevaert
- Stanford Center for Biomedical Informatics Research, Department of Medicine &Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | | | - Amanda Khuong
- Thoracic and GI Oncology Branch, CCR, National Institutes of Health, National Cancer Institute, Bethesda, MD, USA
| | - Chuong D Hoang
- Thoracic and GI Oncology Branch, CCR, National Institutes of Health, National Cancer Institute, Bethesda, MD, USA
| | - Joseph B Shrager
- Thoracic and GI Oncology Branch, CCR, National Institutes of Health, National Cancer Institute, Bethesda, MD, USA
| | - Kirstin C Jensen
- Department of Pathology, Stanford University Medical Center, Stanford, CA, USA.,Pathology and Laboratory Service of Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Gerald J Berry
- Department of Pathology, Stanford University Medical Center, Stanford, CA, USA
| | - H Henry Guo
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Charles Lau
- Department of Radiology, Stanford University, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | | | - Daniel L Rubin
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Sandy Napel
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Ann N Leung
- Department of Radiology, Stanford University, Stanford, CA, USA
| |
Collapse
|
314
|
Lambin P, Zindler J, Vanneste BGL, De Voorde LV, Eekers D, Compter I, Panth KM, Peerlings J, Larue RTHM, Deist TM, Jochems A, Lustberg T, van Soest J, de Jong EEC, Even AJG, Reymen B, Rekers N, van Gisbergen M, Roelofs E, Carvalho S, Leijenaar RTH, Zegers CML, Jacobs M, van Timmeren J, Brouwers P, Lal JA, Dubois L, Yaromina A, Van Limbergen EJ, Berbee M, van Elmpt W, Oberije C, Ramaekers B, Dekker A, Boersma LJ, Hoebers F, Smits KM, Berlanga AJ, Walsh S. Decision support systems for personalized and participative radiation oncology. Adv Drug Deliv Rev 2017; 109:131-153. [PMID: 26774327 DOI: 10.1016/j.addr.2016.01.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/08/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022]
Abstract
A paradigm shift from current population based medicine to personalized and participative medicine is underway. This transition is being supported by the development of clinical decision support systems based on prediction models of treatment outcome. In radiation oncology, these models 'learn' using advanced and innovative information technologies (ideally in a distributed fashion - please watch the animation: http://youtu.be/ZDJFOxpwqEA) from all available/appropriate medical data (clinical, treatment, imaging, biological/genetic, etc.) to achieve the highest possible accuracy with respect to prediction of tumor response and normal tissue toxicity. In this position paper, we deliver an overview of the factors that are associated with outcome in radiation oncology and discuss the methodology behind the development of accurate prediction models, which is a multi-faceted process. Subsequent to initial development/validation and clinical introduction, decision support systems should be constantly re-evaluated (through quality assurance procedures) in different patient datasets in order to refine and re-optimize the models, ensuring the continuous utility of the models. In the reasonably near future, decision support systems will be fully integrated within the clinic, with data and knowledge being shared in a standardized, dynamic, and potentially global manner enabling truly personalized and participative medicine.
Collapse
Affiliation(s)
- Philippe Lambin
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Jaap Zindler
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ben G L Vanneste
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Lien Van De Voorde
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Daniëlle Eekers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Inge Compter
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Kranthi Marella Panth
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jurgen Peerlings
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ruben T H M Larue
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Timo M Deist
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Arthur Jochems
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Tim Lustberg
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Johan van Soest
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Evelyn E C de Jong
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Aniek J G Even
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Bart Reymen
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Nicolle Rekers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Marike van Gisbergen
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Erik Roelofs
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sara Carvalho
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ralph T H Leijenaar
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Catharina M L Zegers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Maria Jacobs
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Janita van Timmeren
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Patricia Brouwers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jonathan A Lal
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ludwig Dubois
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ala Yaromina
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Evert Jan Van Limbergen
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Maaike Berbee
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Cary Oberije
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Bram Ramaekers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Liesbeth J Boersma
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Frank Hoebers
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Kim M Smits
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Adriana J Berlanga
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sean Walsh
- Department of Radiation Oncology (MAASTRO), GROW, School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
315
|
Huynh E, Coroller TP, Narayan V, Agrawal V, Romano J, Franco I, Parmar C, Hou Y, Mak RH, Aerts HJWL. Associations of Radiomic Data Extracted from Static and Respiratory-Gated CT Scans with Disease Recurrence in Lung Cancer Patients Treated with SBRT. PLoS One 2017; 12:e0169172. [PMID: 28046060 PMCID: PMC5207741 DOI: 10.1371/journal.pone.0169172] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 12/13/2016] [Indexed: 02/06/2023] Open
Abstract
Radiomics aims to quantitatively capture the complex tumor phenotype contained in medical images to associate them with clinical outcomes. This study investigates the impact of different types of computed tomography (CT) images on the prognostic performance of radiomic features for disease recurrence in early stage non-small cell lung cancer (NSCLC) patients treated with stereotactic body radiation therapy (SBRT). 112 early stage NSCLC patients treated with SBRT that had static free breathing (FB) and average intensity projection (AIP) images were analyzed. Nineteen radiomic features were selected from each image type (FB or AIP) for analysis based on stability and variance. The selected FB and AIP radiomic feature sets had 6 common radiomic features between both image types and 13 unique features. The prognostic performances of the features for distant metastasis (DM) and locoregional recurrence (LRR) were evaluated using the concordance index (CI) and compared with two conventional features (tumor volume and maximum diameter). P-values were corrected for multiple testing using the false discovery rate procedure. None of the FB radiomic features were associated with DM, however, seven AIP radiomic features, that described tumor shape and heterogeneity, were (CI range: 0.638-0.676). Conventional features from FB images were not associated with DM, however, AIP conventional features were (CI range: 0.643-0.658). Radiomic and conventional multivariate models were compared between FB and AIP images using cross validation. The differences between the models were assessed using a permutation test. AIP radiomic multivariate models (median CI = 0.667) outperformed all other models (median CI range: 0.601-0.630) in predicting DM. None of the imaging features were prognostic of LRR. Therefore, image type impacts the performance of radiomic models in their association with disease recurrence. AIP images contained more information than FB images that were associated with disease recurrence in early stage NSCLC patients treated with SBRT, which suggests that AIP images may potentially be more optimal for the development of an imaging biomarker.
Collapse
Affiliation(s)
- Elizabeth Huynh
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
- * E-mail:
| | - Thibaud P. Coroller
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Vivek Narayan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Vishesh Agrawal
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - John Romano
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Idalid Franco
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Chintan Parmar
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Ying Hou
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Raymond H. Mak
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Hugo J. W. L. Aerts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
- Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States of America
| |
Collapse
|
316
|
Sala E, Mema E, Himoto Y, Veeraraghavan H, Brenton JD, Snyder A, Weigelt B, Vargas HA. Unravelling tumour heterogeneity using next-generation imaging: radiomics, radiogenomics, and habitat imaging. Clin Radiol 2017; 72:3-10. [PMID: 27742105 PMCID: PMC5503113 DOI: 10.1016/j.crad.2016.09.013] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/06/2016] [Accepted: 09/12/2016] [Indexed: 12/18/2022]
Abstract
Tumour heterogeneity in cancers has been observed at the histological and genetic levels, and increased levels of intra-tumour genetic heterogeneity have been reported to be associated with adverse clinical outcomes. This review provides an overview of radiomics, radiogenomics, and habitat imaging, and examines the use of these newly emergent fields in assessing tumour heterogeneity and its implications. It reviews the potential value of radiomics and radiogenomics in assisting in the diagnosis of cancer disease and determining cancer aggressiveness. This review discusses how radiogenomic analysis can be further used to guide treatment therapy for individual tumours by predicting drug response and potential therapy resistance and examines its role in developing radiomics as biomarkers of oncological outcomes. Lastly, it provides an overview of the obstacles in these emergent fields today including reproducibility, need for validation, imaging analysis standardisation, data sharing and clinical translatability and offers potential solutions to these challenges towards the realisation of precision oncology.
Collapse
Affiliation(s)
- E Sala
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - E Mema
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Department of Radiology, New York Presbyterian/Columbia University Medical Center, 622 W 168th St., New York, NY 10032, USA
| | - Y Himoto
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - H Veeraraghavan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - J D Brenton
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - A Snyder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - B Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - H A Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
317
|
Hatt M, Tixier F, Pierce L, Kinahan PE, Le Rest CC, Visvikis D. Characterization of PET/CT images using texture analysis: the past, the present… any future? Eur J Nucl Med Mol Imaging 2017; 44:151-165. [PMID: 27271051 PMCID: PMC5283691 DOI: 10.1007/s00259-016-3427-0] [Citation(s) in RCA: 338] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/18/2016] [Indexed: 02/07/2023]
Abstract
After seminal papers over the period 2009 - 2011, the use of texture analysis of PET/CT images for quantification of intratumour uptake heterogeneity has received increasing attention in the last 4 years. Results are difficult to compare due to the heterogeneity of studies and lack of standardization. There are also numerous challenges to address. In this review we provide critical insights into the recent development of texture analysis for quantifying the heterogeneity in PET/CT images, identify issues and challenges, and offer recommendations for the use of texture analysis in clinical research. Numerous potentially confounding issues have been identified, related to the complex workflow for the calculation of textural features, and the dependency of features on various factors such as acquisition, image reconstruction, preprocessing, functional volume segmentation, and methods of establishing and quantifying correspondences with genomic and clinical metrics of interest. A lack of understanding of what the features may represent in terms of the underlying pathophysiological processes and the variability of technical implementation practices makes comparing results in the literature challenging, if not impossible. Since progress as a field requires pooling results, there is an urgent need for standardization and recommendations/guidelines to enable the field to move forward. We provide a list of correct formulae for usual features and recommendations regarding implementation. Studies on larger cohorts with robust statistical analysis and machine learning approaches are promising directions to evaluate the potential of this approach.
Collapse
Affiliation(s)
- Mathieu Hatt
- INSERM, UMR 1101, LaTIM, University of Brest IBSAM, Brest, France.
| | - Florent Tixier
- Nuclear Medicine, University Hospital, Poitiers, France
- Medical school, EE DACTIM, University of Poitiers, Poitiers, France
| | - Larry Pierce
- Imaging Research Laboratory, University of Washington, Seattle, WA, USA
| | - Paul E Kinahan
- Imaging Research Laboratory, University of Washington, Seattle, WA, USA
| | - Catherine Cheze Le Rest
- Nuclear Medicine, University Hospital, Poitiers, France
- Medical school, EE DACTIM, University of Poitiers, Poitiers, France
| | | |
Collapse
|
318
|
Zhao H, Hua Y, Dai T, He J, Tang M, Fu X, Mao L, Jin H, Qiu Y. Development and validation of a novel predictive scoring model for microvascular invasion in patients with hepatocellular carcinoma. Eur J Radiol 2016; 88:32-40. [PMID: 28189206 DOI: 10.1016/j.ejrad.2016.12.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/23/2016] [Accepted: 12/25/2016] [Indexed: 12/13/2022]
Abstract
PURPOSE Microvascular invasion (MVI) in patients with hepatocellular carcinoma (HCC) cannot be accurately predicted preoperatively. This study aimed to establish a predictive scoring model of MVI in solitary HCC patients without macroscopic vascular invasion. METHODS A total of 309 consecutive HCC patients who underwent curative hepatectomy were divided into the derivation (n=206) and validation cohort (n=103). A predictive scoring model of MVI was established according to the valuable predictors in the derivation cohort based on multivariate logistic regression analysis. The performance of the predictive model was evaluated in the derivation and validation cohorts. RESULTS Preoperative imaging features on CECT, such as intratumoral arteries, non-nodular type of HCC and absence of radiological tumor capsule were independent predictors for MVI. The predictive scoring model was established according to the β coefficients of the 3 predictors. Area under receiver operating characteristic (AUROC) of the predictive scoring model was 0.872 (95% CI, 0.817-0.928) and 0.856 (95% CI, 0.771-0.940) in the derivation and validation cohorts. The positive and negative predictive values were 76.5% and 88.0% in the derivation cohort and 74.4% and 88.3% in the validation cohort. The performance of the model was similar between the patients with tumor size ≤5cm and >5cm in AUROC (P=0.910). CONCLUSIONS The predictive scoring model based on intratumoral arteries, non-nodular type of HCC, and absence of the radiological tumor capsule on preoperative CECT is of great value in the prediction of MVI regardless of tumor size.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Hepatopancreatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China; Department of Hepatopancreatobiliary Surgery, Nanjing Medical University Affiliated Wuxi Second People's Hospital, Wuxi, Jiangsu, China
| | - Ye Hua
- Department of Neurology, Nanjing Medical University Affiliated Wuxi Second People's Hospital, Wuxi, Jiangsu, China
| | - Tu Dai
- Department of Hepatopancreatobiliary Surgery, Nanjing Medical University Affiliated Wuxi Second People's Hospital, Wuxi, Jiangsu, China
| | - Jian He
- Department of Radiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Min Tang
- Department of Radiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Xu Fu
- Department of Hepatopancreatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liang Mao
- Department of Hepatopancreatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huihan Jin
- Department of Hepatopancreatobiliary Surgery, Nanjing Medical University Affiliated Wuxi Second People's Hospital, Wuxi, Jiangsu, China.
| | - Yudong Qiu
- Department of Hepatopancreatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
319
|
Hesketh RL, Zhu AX, Oklu R. Radiomics and circulating tumor cells: personalized care in hepatocellular carcinoma? Diagn Interv Radiol 2016; 21:78-84. [PMID: 25430530 DOI: 10.5152/dir.2014.14237] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Personalized care in oncology is expected to significantly improve morbidity and mortality, facilitated by our increasing understanding of the molecular mechanisms driving tumors and the ability to target those drivers. Hepatocellular carcinoma has a very high mortality to incidence ratio despite localized disease being curable, emphasizing the importance of early diagnosis. Radiomics, the use of imaging technology to extrapolate molecular tumor data, and the detection of circulating tumor cells (CTCs) are two new technologies that could be incorporated into the clinical setting with relative ease. Here we discuss the molecular mechanisms leading to the development of hepatocellular carcinoma focusing on the latest developments in liver magnetic resonance imaging, CTC, and radiomic technology and their potential to improve diagnosis, staging, and therapy.
Collapse
|
320
|
Larue RTHM, Defraene G, De Ruysscher D, Lambin P, van Elmpt W. Quantitative radiomics studies for tissue characterization: a review of technology and methodological procedures. Br J Radiol 2016; 90:20160665. [PMID: 27936886 PMCID: PMC5685111 DOI: 10.1259/bjr.20160665] [Citation(s) in RCA: 240] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Quantitative analysis of tumour characteristics based on medical imaging is an emerging field of research. In recent years, quantitative imaging features derived from CT, positron emission tomography and MR scans were shown to be of added value in the prediction of outcome parameters in oncology, in what is called the radiomics field. However, results might be difficult to compare owing to a lack of standardized methodologies to conduct quantitative image analyses. In this review, we aim to present an overview of the current challenges, technical routines and protocols that are involved in quantitative imaging studies. The first issue that should be overcome is the dependency of several features on the scan acquisition and image reconstruction parameters. Adopting consistent methods in the subsequent target segmentation step is evenly crucial. To further establish robust quantitative image analyses, standardization or at least calibration of imaging features based on different feature extraction settings is required, especially for texture- and filter-based features. Several open-source and commercial software packages to perform feature extraction are currently available, all with slightly different functionalities, which makes benchmarking quite challenging. The number of imaging features calculated is typically larger than the number of patients studied, which emphasizes the importance of proper feature selection and prediction model-building routines to prevent overfitting. Even though many of these challenges still need to be addressed before quantitative imaging can be brought into daily clinical practice, radiomics is expected to be a critical component for the integration of image-derived information to personalize treatment in the future.
Collapse
Affiliation(s)
- Ruben T H M Larue
- 1 Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Gilles Defraene
- 2 Department of Oncology, Experimental Radiation Oncology, University of Leuven, Leuven, Belgium
| | - Dirk De Ruysscher
- 1 Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Philippe Lambin
- 1 Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Wouter van Elmpt
- 1 Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
321
|
iTRAQ-Based Proteomics Identification of Serum Biomarkers of Two Chronic Hepatitis B Subtypes Diagnosed by Traditional Chinese Medicine. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3290260. [PMID: 28025641 PMCID: PMC5153474 DOI: 10.1155/2016/3290260] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/24/2016] [Indexed: 02/05/2023]
Abstract
Background. Chronic infection with hepatitis B virus (HBV) is a leading cause of cirrhosis and hepatocellular carcinoma. By traditional Chinese medicine (TCM) pattern classification, damp heat stasis in the middle-jiao (DHSM) and liver Qi stagnation and spleen deficiency (LSSD) are two most common subtypes of CHB. Results. In this study, we employed iTRAQ proteomics technology to identify potential serum protein biomarkers in 30 LSSD-CHB and 30 DHSM-CHB patients. Of the total 842 detected proteins, 273 and 345 were differentially expressed in LSSD-CHB and DHSM-CHB patients compared to healthy controls, respectively. LSSD-CHB and DHSM-CHB shared 142 upregulated and 84 downregulated proteins, of which several proteins have been reported to be candidate biomarkers, including immunoglobulin (Ig) related proteins, complement components, apolipoproteins, heat shock proteins, insulin-like growth factor binding protein, and alpha-2-macroglobulin. In addition, we identified that proteins might be potential biomarkers to distinguish LSSD-CHB from DHSM-CHB, such as A0A0A0MS51_HUMAN (gelsolin), PON3_HUMAN, Q96K68_HUMAN, and TRPM8_HUMAN that were differentially expressed exclusively in LSSD-CHB patients and A0A087WT59_HUMAN (transthyretin), ITIH1_HUMAN, TSP1_HUMAN, CO5_HUMAN, and ALBU_HUMAN that were differentially expressed specifically in DHSM-CHB patients. Conclusion. This is the first time to report serum proteins in CHB subtype patients. Our findings provide potential biomarkers can be used for LSSD-CHB and DHSM-CHB.
Collapse
|
322
|
Bashir U, Siddique MM, Mclean E, Goh V, Cook GJ. Imaging Heterogeneity in Lung Cancer: Techniques, Applications, and Challenges. AJR Am J Roentgenol 2016; 207:534-43. [PMID: 27305342 DOI: 10.2214/ajr.15.15864] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Texture analysis involves the mathematic processing of medical images to derive sets of numeric quantities that measure heterogeneity. Studies on lung cancer have shown that texture analysis may have a role in characterizing tumors and predicting patient outcome. This article outlines the mathematic basis of and the most recent literature on texture analysis in lung cancer imaging. We also describe the challenges facing the clinical implementation of texture analysis. CONCLUSION Texture analysis of lung cancer images has been applied successfully to FDG PET and CT scans. Different texture parameters have been shown to be predictive of the nature of disease and of patient outcome. In general, it appears that more heterogeneous tumors on imaging tend to be more aggressive and to be associated with poorer outcomes and that tumor heterogeneity on imaging decreases with treatment. Despite these promising results, there is a large variation in the reported data and strengths of association.
Collapse
Affiliation(s)
- Usman Bashir
- 1 Department of Cancer Imaging, Division of Imaging Sciences and Biomedical Engineering, King's College London, London SE1 7EH, UK
| | - Muhammad Musib Siddique
- 1 Department of Cancer Imaging, Division of Imaging Sciences and Biomedical Engineering, King's College London, London SE1 7EH, UK
| | - Emma Mclean
- 2 Department of Histopathology, Guy's and St. Thomas' Hospitals, London, UK
| | - Vicky Goh
- 1 Department of Cancer Imaging, Division of Imaging Sciences and Biomedical Engineering, King's College London, London SE1 7EH, UK
- 3 Department of Radiology, Guy's and St. Thomas' Hospitals, London, UK
| | - Gary J Cook
- 1 Department of Cancer Imaging, Division of Imaging Sciences and Biomedical Engineering, King's College London, London SE1 7EH, UK
- 4 PET Imaging Centre, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| |
Collapse
|
323
|
Joint analysis of multiple high-dimensional data types using sparse matrix approximations of rank-1 with applications to ovarian and liver cancer. BioData Min 2016; 9:24. [PMID: 27478503 PMCID: PMC4966782 DOI: 10.1186/s13040-016-0103-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 07/05/2016] [Indexed: 12/15/2022] Open
Abstract
Background Technological advances enable the cost-effective acquisition of Multi-Modal Data Sets (MMDS) composed of measurements for multiple, high-dimensional data types obtained from a common set of bio-samples. The joint analysis of the data matrices associated with the different data types of a MMDS should provide a more focused view of the biology underlying complex diseases such as cancer that would not be apparent from the analysis of a single data type alone. As multi-modal data rapidly accumulate in research laboratories and public databases such as The Cancer Genome Atlas (TCGA), the translation of such data into clinically actionable knowledge has been slowed by the lack of computational tools capable of analyzing MMDSs. Here, we describe the Joint Analysis of Many Matrices by ITeration (JAMMIT) algorithm that jointly analyzes the data matrices of a MMDS using sparse matrix approximations of rank-1. Methods The JAMMIT algorithm jointly approximates an arbitrary number of data matrices by rank-1 outer-products composed of “sparse” left-singular vectors (eigen-arrays) that are unique to each matrix and a right-singular vector (eigen-signal) that is common to all the matrices. The non-zero coefficients of the eigen-arrays identify small subsets of variables for each data type (i.e., signatures) that in aggregate, or individually, best explain a dominant eigen-signal defined on the columns of the data matrices. The approximation is specified by a single “sparsity” parameter that is selected based on false discovery rate estimated by permutation testing. Multiple signals of interest in a given MDDS are sequentially detected and modeled by iterating JAMMIT on “residual” data matrices that result from a given sparse approximation. Results We show that JAMMIT outperforms other joint analysis algorithms in the detection of multiple signatures embedded in simulated MDDS. On real multimodal data for ovarian and liver cancer we show that JAMMIT identified multi-modal signatures that were clinically informative and enriched for cancer-related biology. Conclusions Sparse matrix approximations of rank-1 provide a simple yet effective means of jointly reducing multiple, big data types to a small subset of variables that characterize important clinical and/or biological attributes of the bio-samples from which the data were acquired. Electronic supplementary material The online version of this article (doi:10.1186/s13040-016-0103-7) contains supplementary material, which is available to authorized users.
Collapse
|
324
|
Stoyanova R, Takhar M, Tschudi Y, Ford JC, Solórzano G, Erho N, Balagurunathan Y, Punnen S, Davicioni E, Gillies RJ, Pollack A. Prostate cancer radiomics and the promise of radiogenomics. Transl Cancer Res 2016; 5:432-447. [PMID: 29188191 DOI: 10.21037/tcr.2016.06.20] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prostate cancer exhibits intra-tumoral heterogeneity that we hypothesize to be the leading confounding factor contributing to the underperformance of the current pre-treatment clinical-pathological and genomic assessment. These limitations impose an urgent need to develop better computational tools to identify men with low risk of prostate cancer versus others that may be at risk for developing metastatic cancer. The patient stratification will directly translate to patient treatments, wherein decisions regarding active surveillance or intensified therapy are made. Multiparametric MRI (mpMRI) provides the platform to investigate tumor heterogeneity by mapping the individual tumor habitats. We hypothesize that quantitative assessment (radiomics) of these habitats results in distinct combinations of descriptors that reveal regions with different physiologies and phenotypes. Radiogenomics, a discipline connecting tumor morphology described by radiomic and its genome described by the genomic data, has the potential to derive "radio phenotypes" that both correlate to and complement existing validated genomic risk stratification biomarkers. In this article we first describe the radiomic pipeline, tailored for analysis of prostate mpMRI, and in the process we introduce our particular implementations of radiomics modules. We also summarize the efforts in the radiomics field related to prostate cancer diagnosis and assessment of aggressiveness. Finally, we describe our results from radiogenomic analysis, based on mpMRI-Ultrasound (MRI-US) biopsies and discuss the potential of future applications of this technique. The mpMRI radiomics data indicate that the platform would significantly improve the biopsy targeting of prostate habitats through better recognition of indolent versus aggressive disease, thereby facilitating a more personalized approach to prostate cancer management. The expectation to non-invasively identify habitats with high probability of housing aggressive cancers would result in directed biopsies that are more informative and actionable. Conversely, providing evidence for lack of disease would reduce the incidence of non-informative biopsies. In radiotherapy of prostate cancer, dose escalation has been shown to reduce biochemical failure. Dose escalation only to determinate prostate habitats has the potential to improve tumor control with less toxicity than when the entire prostate is dose escalated.
Collapse
Affiliation(s)
- Radka Stoyanova
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mandeep Takhar
- Research and Development, GenomeDx Biosciences, Vancouver, BC, Canada
| | - Yohann Tschudi
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - John C Ford
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gabriel Solórzano
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nicholas Erho
- Research and Development, GenomeDx Biosciences, Vancouver, BC, Canada
| | | | - Sanoj Punnen
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Elai Davicioni
- Research and Development, GenomeDx Biosciences, Vancouver, BC, Canada
| | - Robert J Gillies
- Cancer Imaging and Metabolism, Moffitt Cancer Center, Tampa, FL, USA
| | - Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
325
|
Tang H, Bai HX, Su C, Lee AM, Yang L. The effect of cirrhosis on radiogenomic biomarker's ability to predict microvascular invasion and outcome in hepatocellular carcinoma. Hepatology 2016; 64:691-2. [PMID: 27113641 DOI: 10.1002/hep.28620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Haiyun Tang
- Department of Radiology, The First Xiangya Hospital, Central South University, Changsha, China
| | - Harrison X Bai
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Chang Su
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
| | - Ashley M Lee
- Mallinckrodt Institute of Radiology, Washington University in Saint Louis, St. Louis, MO
| | - Li Yang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
326
|
Abstract
OBJECTIVES The purpose of this study was to determine the significance of interscanner variability in CT image radiomics studies. MATERIALS AND METHODS We compared the radiomics features calculated for non-small cell lung cancer (NSCLC) tumors from 20 patients with those calculated for 17 scans of a specially designed radiomics phantom. The phantom comprised 10 cartridges, each filled with different materials to produce a wide range of radiomics feature values. The scans were acquired using General Electric, Philips, Siemens, and Toshiba scanners from 4 medical centers using their routine thoracic imaging protocol. The radiomics feature studied included the mean and standard deviations of the CT numbers as well as textures derived from the neighborhood gray-tone difference matrix. To quantify the significance of the interscanner variability, we introduced the metric feature noise. To look for patterns in the scans, we performed hierarchical clustering for each cartridge. RESULTS The mean CT numbers for the 17 CT scans of the phantom cartridges spanned from -864 to 652 Hounsfield units compared with a span of -186 to 35 Hounsfield units for the CT scans of the NSCLC tumors, showing that the phantom's dynamic range includes that of the tumors. The interscanner variability of the feature values depended on both the cartridge material and the feature, and the variability was large relative to the interpatient variability in the NSCLC tumors for some features. The feature interscanner noise was greatest for busyness and least for texture strength. Hierarchical clustering produced different clusters of the phantom scans for each cartridge, although there was some consistent clustering by scanner manufacturer. CONCLUSIONS The variability in the values of radiomics features calculated on CT images from different CT scanners can be comparable to the variability in these features found in CT images of NSCLC tumors. These interscanner differences should be considered, and their effects should be minimized in future radiomics studies.
Collapse
|
327
|
Huynh E, Coroller TP, Narayan V, Agrawal V, Hou Y, Romano J, Franco I, Mak RH, Aerts HJWL. CT-based radiomic analysis of stereotactic body radiation therapy patients with lung cancer. Radiother Oncol 2016; 120:258-66. [PMID: 27296412 DOI: 10.1016/j.radonc.2016.05.024] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 05/23/2016] [Accepted: 05/24/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Radiomics uses a large number of quantitative imaging features that describe the tumor phenotype to develop imaging biomarkers for clinical outcomes. Radiomic analysis of pre-treatment computed-tomography (CT) scans was investigated to identify imaging predictors of clinical outcomes in early stage non-small cell lung cancer (NSCLC) patients treated with stereotactic body radiation therapy (SBRT). MATERIALS AND METHODS CT images of 113 stage I-II NSCLC patients treated with SBRT were analyzed. Twelve radiomic features were selected based on stability and variance. The association of features with clinical outcomes and their prognostic value (using the concordance index (CI)) was evaluated. Radiomic features were compared with conventional imaging metrics (tumor volume and diameter) and clinical parameters. RESULTS Overall survival was associated with two conventional features (volume and diameter) and two radiomic features (LoG 3D run low gray level short run emphasis and stats median). One radiomic feature (Wavelet LLH stats range) was significantly prognostic for distant metastasis (CI=0.67, q-value<0.1), while none of the conventional and clinical parameters were. Three conventional and four radiomic features were prognostic for overall survival. CONCLUSION This exploratory analysis demonstrates that radiomic features have potential to be prognostic for some outcomes that conventional imaging metrics cannot predict in SBRT patients.
Collapse
Affiliation(s)
- Elizabeth Huynh
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.
| | - Thibaud P Coroller
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Vivek Narayan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Vishesh Agrawal
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Ying Hou
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - John Romano
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Idalid Franco
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Raymond H Mak
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Hugo J W L Aerts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
328
|
Wu C, Chen J, Kim J, Pan W. An adaptive association test for microbiome data. Genome Med 2016; 8:56. [PMID: 27198579 PMCID: PMC4872356 DOI: 10.1186/s13073-016-0302-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/12/2016] [Indexed: 02/07/2023] Open
Abstract
There is increasing interest in investigating how the compositions of microbial communities are associated with human health and disease. Although existing methods have identified many associations, a proper choice of a phylogenetic distance is critical for the power of these methods. To assess an overall association between the composition of a microbial community and an outcome of interest, we present a novel multivariate testing method called aMiSPU, that is joint and highly adaptive over all observed taxa and thus high powered across various scenarios, alleviating the issue with the choice of a phylogenetic distance. Our simulations and real-data analyses demonstrated that the aMiSPU test was often more powerful than several competing methods while correctly controlling type I error rates. The R package MiSPU is available at https://github.com/ChongWu-Biostat/MiSPU
and CRAN.
Collapse
Affiliation(s)
- Chong Wu
- Division of Biostatistics, University of Minnesota, 420 Delaware St. SE, Minneapolis, 55455, USA
| | - Jun Chen
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First St. SW, Rochester, 55905, USA
| | - Junghi Kim
- Division of Biostatistics, University of Minnesota, 420 Delaware St. SE, Minneapolis, 55455, USA
| | - Wei Pan
- Division of Biostatistics, University of Minnesota, 420 Delaware St. SE, Minneapolis, 55455, USA.
| |
Collapse
|
329
|
Liu TY, Dodson AE, Terhorst J, Song YS, Rine J. Riches of phenotype computationally extracted from microbial colonies. Proc Natl Acad Sci U S A 2016; 113:E2822-31. [PMID: 27140647 PMCID: PMC4878510 DOI: 10.1073/pnas.1523295113] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The genetic, epigenetic, and physiological differences among cells in clonal microbial colonies are underexplored opportunities for discovery. A recently developed genetic assay reveals that transient losses of heterochromatic repression, a heritable form of gene silencing, occur throughout the growth of Saccharomyces colonies. This assay requires analyzing two-color fluorescence patterns in yeast colonies, which is qualitatively appealing but quantitatively challenging. In this paper, we developed a suite of automated image processing, visualization, and classification algorithms (MORPHE) that facilitated the analysis of heterochromatin dynamics in the context of colonial growth and that can be broadly adapted to many colony-based assays in Saccharomyces and other microbes. Using the features that were automatically extracted from fluorescence images, our classification method distinguished loss-of-silencing patterns between mutants and wild type with unprecedented precision. Application of MORPHE revealed subtle but significant differences in the stability of heterochromatic repression between various environmental conditions, revealed that haploid cells experienced higher rates of silencing loss than diploids, and uncovered the unexpected contribution of a sirtuin to heterochromatin dynamics.
Collapse
Affiliation(s)
- Tzu-Yu Liu
- Department of Mathematics and Department of Biology, University of Pennsylvania, Philadelphia, PA 19104; Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720
| | - Anne E Dodson
- Department of Molecular and Cell Biology and California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
| | - Jonathan Terhorst
- Department of Statistics, University of California, Berkeley, CA 94720
| | - Yun S Song
- Department of Mathematics and Department of Biology, University of Pennsylvania, Philadelphia, PA 19104; Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720; Department of Statistics, University of California, Berkeley, CA 94720; Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Jasper Rine
- Department of Molecular and Cell Biology and California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720;
| |
Collapse
|
330
|
Zhang L, Fried DV, Fave XJ, Hunter LA, Yang J, Court LE. IBEX: an open infrastructure software platform to facilitate collaborative work in radiomics. Med Phys 2016; 42:1341-53. [PMID: 25735289 DOI: 10.1118/1.4908210] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Radiomics, which is the high-throughput extraction and analysis of quantitative image features, has been shown to have considerable potential to quantify the tumor phenotype. However, at present, a lack of software infrastructure has impeded the development of radiomics and its applications. Therefore, the authors developed the imaging biomarker explorer (IBEX), an open infrastructure software platform that flexibly supports common radiomics workflow tasks such as multimodality image data import and review, development of feature extraction algorithms, model validation, and consistent data sharing among multiple institutions. METHODS The IBEX software package was developed using the MATLAB and c/c++ programming languages. The software architecture deploys the modern model-view-controller, unit testing, and function handle programming concepts to isolate each quantitative imaging analysis task, to validate if their relevant data and algorithms are fit for use, and to plug in new modules. On one hand, IBEX is self-contained and ready to use: it has implemented common data importers, common image filters, and common feature extraction algorithms. On the other hand, IBEX provides an integrated development environment on top of MATLAB and c/c++, so users are not limited to its built-in functions. In the IBEX developer studio, users can plug in, debug, and test new algorithms, extending IBEX's functionality. IBEX also supports quality assurance for data and feature algorithms: image data, regions of interest, and feature algorithm-related data can be reviewed, validated, and/or modified. More importantly, two key elements in collaborative workflows, the consistency of data sharing and the reproducibility of calculation result, are embedded in the IBEX workflow: image data, feature algorithms, and model validation including newly developed ones from different users can be easily and consistently shared so that results can be more easily reproduced between institutions. RESULTS Researchers with a variety of technical skill levels, including radiation oncologists, physicists, and computer scientists, have found the IBEX software to be intuitive, powerful, and easy to use. IBEX can be run at any computer with the windows operating system and 1GB RAM. The authors fully validated the implementation of all importers, preprocessing algorithms, and feature extraction algorithms. Windows version 1.0 beta of stand-alone IBEX and IBEX's source code can be downloaded. CONCLUSIONS The authors successfully implemented IBEX, an open infrastructure software platform that streamlines common radiomics workflow tasks. Its transparency, flexibility, and portability can greatly accelerate the pace of radiomics research and pave the way toward successful clinical translation.
Collapse
Affiliation(s)
- Lifei Zhang
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - David V Fried
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 and The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas 77030
| | - Xenia J Fave
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 and The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas 77030
| | - Luke A Hunter
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 and The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas 77030
| | - Jinzhong Yang
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Laurence E Court
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030 and The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas 77030
| |
Collapse
|
331
|
Rusu M, Golden T, Wang H, Gow A, Madabhushi A. Framework for 3D histologic reconstruction and fusion with in vivo MRI: Preliminary results of characterizing pulmonary inflammation in a mouse model. Med Phys 2016; 42:4822-32. [PMID: 26233209 DOI: 10.1118/1.4923161] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE Pulmonary inflammation is associated with a variety of diseases. Assessing pulmonary inflammation on in vivo imaging may facilitate the early detection and treatment of lung diseases. Although routinely used in thoracic imaging, computed tomography has thus far not been compellingly shown to characterize inflammation in vivo. Alternatively, magnetic resonance imaging (MRI) is a nonionizing radiation technique to better visualize and characterize pulmonary tissue. Prior to routine adoption of MRI for early characterization of inflammation in humans, a rigorous and quantitative characterization of the utility of MRI to identify inflammation is required. Such characterization may be achieved by considering ex vivo histology as the ground truth, since it enables the definitive spatial assessment of inflammation. In this study, the authors introduce a novel framework to integrate 2D histology, ex vivo and in vivo imaging to enable the mapping of the extent of disease from ex vivo histology onto in vivo imaging, with the goal of facilitating computerized feature analysis and interrogation of disease appearance on in vivo imaging. The authors' framework was evaluated in a preclinical preliminary study aimed to identify computer extracted features on in vivo MRI associated with chronic pulmonary inflammation. METHODS The authors' image analytics framework first involves reconstructing the histologic volume in 3D from individual histology slices. Second, the authors map the disease ground truth onto in vivo MRI via coregistration with 3D histology using the ex vivo lung MRI as a conduit. Finally, computerized feature analysis of the disease extent is performed to identify candidate in vivo imaging signatures of disease presence and extent. RESULTS The authors evaluated the framework by assessing the quality of the 3D histology reconstruction and the histology-MRI fusion, in the context of an initial use case involving characterization of chronic inflammation in a mouse model. The authors' evaluation considered three mice, two with an inflammation phenotype and one control. The authors' iterative 3D histology reconstruction yielded a 70.1% ± 2.7% overlap with the ex vivo MRI volume. Across a total of 17 anatomic landmarks manually delineated at the division of airways, the target registration error between the ex vivo MRI and 3D histology reconstruction was 0.85 ± 0.44 mm, suggesting that a good alignment of the ex vivo 3D histology and ex vivo MRI had been achieved. The 3D histology-in vivo MRI coregistered volumes resulted in an overlap of 73.7% ± 0.9%. Preliminary computerized feature analysis was performed on an additional four control mice, for a total of seven mice considered in this study. Gabor texture filters appeared to best capture differences between the inflamed and noninflamed regions on MRI. CONCLUSIONS The authors' 3D histology reconstruction and multimodal registration framework were successfully employed to reconstruct the histology volume of the lung and fuse it with in vivo MRI to create a ground truth map for inflammation on in vivo MRI. The analytic platform presented here lays the framework for a rigorous validation of the identified imaging features for chronic lung inflammation on MRI in a large prospective cohort.
Collapse
Affiliation(s)
- Mirabela Rusu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106
| | - Thea Golden
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey 08854
| | - Haibo Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106
| | - Andrew Gow
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey 08854
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106
| |
Collapse
|
332
|
Yamamoto S, Huang D, Du L, Korn RL, Jamshidi N, Burnette BL, Kuo MD. Radiogenomic Analysis Demonstrates Associations between (18)F-Fluoro-2-Deoxyglucose PET, Prognosis, and Epithelial-Mesenchymal Transition in Non-Small Cell Lung Cancer. Radiology 2016; 280:261-70. [PMID: 27082783 DOI: 10.1148/radiol.2016160259] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Purpose To investigate whether non-small cell lung cancer (NSCLC) tumors that express high normalized maximum standardized uptake value (SUVmax) are associated with a more epithelial-mesenchymal transition (EMT)-like phenotype. Materials and Methods In this institutional review board-approved study, a public NSCLC data set that contained fluorine 18 ((18)F) fluoro-2-deoxyglucose positron emission tomography (PET) and messenger RNA expression profile data (n = 26) was obtained, and patients were categorized on the basis of measured normalized SUVmax values. Significance analysis of microarrays was then used to create a radiogenomic signature. The prognostic ability of this signature was assessed in a second independent data set that consisted of clinical and messenger RNA expression data (n = 166). Signature concordance with EMT was evaluated by means of validation in a publicly available cell line data set. Finally, by establishing an in vitro EMT lung cancer cell line model, an attempt was made to substantiate the radiogenomic signature with quantitative polymerase chain reaction, and functional assays were performed, including Western blot, cell migration, glucose transporter, and hexokinase assays (paired t test), as well as pharmacologic assays against chemotherapeutic agents (half-maximal effective concentration). Results Differential expression analysis yielded a 14-gene radiogenomic signature (P < .05, false discovery rate [FDR] < 0.20), which was confirmed to have differences in disease-specific survival (log-rank test, P = .01). This signature also significantly overlapped with published EMT cell line gene expression data (P < .05, FDR < 0.20). Finally, an EMT cell line model was established, and cells that had undergone EMT differentially expressed this signature and had significantly different EMT protein expression (P < .05, FDR < 0.20), cell migration, glucose uptake, and hexokinase activity (paired t test, P < .05). Cells that had undergone EMT also had enhanced chemotherapeutic resistance, with a higher half-maximal effective concentration than that of cells that had not undergone EMT (P < .05). Conclusion Integrative radiogenomic analysis demonstrates an association between increased normalized (18)F fluoro-2-deoxyglucose PET SUVmax, outcome, and EMT in NSCLC. (©) RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Shota Yamamoto
- From the Department of Radiology, The David Geffen School of Medicine at University of California-Los Angeles (UCLA), 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721 (S.Y., D.H., L.D., N.J., B.L.B., M.D.K.); Department of Bioengineering, UCLA, Los Angeles, Calif (M.D.K.); and Scottsdale Medical Imaging, Scottsdale, Ariz (R.L.K.)
| | - Danshan Huang
- From the Department of Radiology, The David Geffen School of Medicine at University of California-Los Angeles (UCLA), 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721 (S.Y., D.H., L.D., N.J., B.L.B., M.D.K.); Department of Bioengineering, UCLA, Los Angeles, Calif (M.D.K.); and Scottsdale Medical Imaging, Scottsdale, Ariz (R.L.K.)
| | - Liutao Du
- From the Department of Radiology, The David Geffen School of Medicine at University of California-Los Angeles (UCLA), 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721 (S.Y., D.H., L.D., N.J., B.L.B., M.D.K.); Department of Bioengineering, UCLA, Los Angeles, Calif (M.D.K.); and Scottsdale Medical Imaging, Scottsdale, Ariz (R.L.K.)
| | - Ronald L Korn
- From the Department of Radiology, The David Geffen School of Medicine at University of California-Los Angeles (UCLA), 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721 (S.Y., D.H., L.D., N.J., B.L.B., M.D.K.); Department of Bioengineering, UCLA, Los Angeles, Calif (M.D.K.); and Scottsdale Medical Imaging, Scottsdale, Ariz (R.L.K.)
| | - Neema Jamshidi
- From the Department of Radiology, The David Geffen School of Medicine at University of California-Los Angeles (UCLA), 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721 (S.Y., D.H., L.D., N.J., B.L.B., M.D.K.); Department of Bioengineering, UCLA, Los Angeles, Calif (M.D.K.); and Scottsdale Medical Imaging, Scottsdale, Ariz (R.L.K.)
| | - Barry L Burnette
- From the Department of Radiology, The David Geffen School of Medicine at University of California-Los Angeles (UCLA), 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721 (S.Y., D.H., L.D., N.J., B.L.B., M.D.K.); Department of Bioengineering, UCLA, Los Angeles, Calif (M.D.K.); and Scottsdale Medical Imaging, Scottsdale, Ariz (R.L.K.)
| | - Michael D Kuo
- From the Department of Radiology, The David Geffen School of Medicine at University of California-Los Angeles (UCLA), 10833 LeConte Ave, Box 951721, CHS 17-135, Los Angeles, CA 90095-1721 (S.Y., D.H., L.D., N.J., B.L.B., M.D.K.); Department of Bioengineering, UCLA, Los Angeles, Calif (M.D.K.); and Scottsdale Medical Imaging, Scottsdale, Ariz (R.L.K.)
| |
Collapse
|
333
|
Rao A, Rao G, Gutman DA, Flanders AE, Hwang SN, Rubin DL, Colen RR, Zinn PO, Jain R, Wintermark M, Kirby JS, Jaffe CC, Freymann J. A combinatorial radiographic phenotype may stratify patient survival and be associated with invasion and proliferation characteristics in glioblastoma. J Neurosurg 2016; 124:1008-17. [PMID: 26473782 PMCID: PMC4990448 DOI: 10.3171/2015.4.jns142732] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Individual MRI characteristics (e.g., volume) are routinely used to identify survival-associated phenotypes for glioblastoma (GBM). This study investigated whether combinations of MRI features can also stratify survival. Furthermore, the molecular differences between phenotype-induced groups were investigated. METHODS Ninety-two patients with imaging, molecular, and survival data from the TCGA (The Cancer Genome Atlas)-GBM collection were included in this study. For combinatorial phenotype analysis, hierarchical clustering was used. Groups were defined based on a cutpoint obtained via tree-based partitioning. Furthermore, differential expression analysis of microRNA (miRNA) and mRNA expression data was performed using GenePattern Suite. Functional analysis of the resulting genes and miRNAs was performed using Ingenuity Pathway Analysis. Pathway analysis was performed using Gene Set Enrichment Analysis. RESULTS Clustering analysis reveals that image-based grouping of the patients is driven by 3 features: volume-class, hemorrhage, and T1/FLAIR-envelope ratio. A combination of these features stratifies survival in a statistically significant manner. A cutpoint analysis yields a significant survival difference in the training set (median survival difference: 12 months, p = 0.004) as well as a validation set (p = 0.0001). Specifically, a low value for any of these 3 features indicates favorable survival characteristics. Differential expression analysis between cutpoint-induced groups suggests that several immune-associated (natural killer cell activity, T-cell lymphocyte differentiation) and metabolism-associated (mitochondrial activity, oxidative phosphorylation) pathways underlie the transition of this phenotype. Integrating data for mRNA and miRNA suggests the roles of several genes regulating proliferation and invasion. CONCLUSIONS A 3-way combination of MRI phenotypes may be capable of stratifying survival in GBM. Examination of molecular processes associated with groups created by this combinatorial phenotype suggests the role of biological processes associated with growth and invasion characteristics.
Collapse
Affiliation(s)
- Arvind Rao
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ganesh Rao
- Department of Neurosurgery, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - David A. Gutman
- Department of Biomedical Informatics, Emory University, Atlanta, Georgia
| | - Adam E. Flanders
- Department of Radiology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Scott N. Hwang
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Daniel L. Rubin
- Department of Radiology, Stanford University, Stanford, California
| | - Rivka R. Colen
- Department of Diagnostic Radiology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Pascal O. Zinn
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Rajan Jain
- Department of Radiology, New York University School of Medicine, New York, New York
| | - Max Wintermark
- Department of Interventional Neuroradiology, Stanford Medical Center, Stanford, California
| | - Justin S. Kirby
- Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland
| | - C. Carl Jaffe
- Department of Radiology, Boston University School of Medicine, Boston, Massachusetts
| | - John Freymann
- Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland
| |
Collapse
|
334
|
Pseudo progression identification of glioblastoma with dictionary learning. Comput Biol Med 2016; 73:94-101. [PMID: 27100835 DOI: 10.1016/j.compbiomed.2016.03.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/28/2016] [Accepted: 03/30/2016] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Although the use of temozolomide in chemoradiotherapy is effective, the challenging clinical problem of pseudo progression has been raised in brain tumor treatment. This study aims to distinguish pseudo progression from true progression. MATERIALS AND METHODS Between 2000 and 2012, a total of 161 patients with glioblastoma multiforme (GBM) were treated with chemoradiotherapy at our hospital. Among the patients, 79 had their diffusion tensor imaging (DTI) data acquired at the earliest diagnosed date of pseudo progression or true progression, and 23 had both DTI data and genomic data. Clinical records of all patients were kept in good condition. Volumetric fractional anisotropy (FA) images obtained from the DTI data were decomposed into a sequence of sparse representations. Then, a feature selection algorithm was applied to extract the critical features from the feature matrix to reduce the size of the feature matrix and to improve the classification accuracy. RESULTS The proposed approach was validated using the 79 samples with clinical DTI data. Satisfactory results were obtained under different experimental conditions. The area under the receiver operating characteristic (ROC) curve (AUC) was 0.87 for a given dictionary with 1024 atoms. For the subgroup of 23 samples, genomics data analysis was also performed. Results implied further perspective on pseudo progression classification. CONCLUSIONS The proposed method can determine pseudo progression and true progression with improved accuracy. Laboring segmentation is no longer necessary because this skillfully designed method is not sensitive to tumor location.
Collapse
|
335
|
Wu W, Parmar C, Grossmann P, Quackenbush J, Lambin P, Bussink J, Mak R, Aerts HJWL. Exploratory Study to Identify Radiomics Classifiers for Lung Cancer Histology. Front Oncol 2016; 6:71. [PMID: 27064691 PMCID: PMC4811956 DOI: 10.3389/fonc.2016.00071] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 03/14/2016] [Indexed: 01/05/2023] Open
Abstract
Background Radiomics can quantify tumor phenotypic characteristics non-invasively by applying feature algorithms to medical imaging data. In this study of lung cancer patients, we investigated the association between radiomic features and the tumor histologic subtypes (adenocarcinoma and squamous cell carcinoma). Furthermore, in order to predict histologic subtypes, we employed machine-learning methods and independently evaluated their prediction performance. Methods Two independent radiomic cohorts with a combined size of 350 patients were included in our analysis. A total of 440 radiomic features were extracted from the segmented tumor volumes of pretreatment CT images. These radiomic features quantify tumor phenotypic characteristics on medical images using tumor shape and size, intensity statistics, and texture. Univariate analysis was performed to assess each feature’s association with the histological subtypes. In our multivariate analysis, we investigated 24 feature selection methods and 3 classification methods for histology prediction. Multivariate models were trained on the training cohort and their performance was evaluated on the independent validation cohort using the area under ROC curve (AUC). Histology was determined from surgical specimen. Results In our univariate analysis, we observed that fifty-three radiomic features were significantly associated with tumor histology. In multivariate analysis, feature selection methods ReliefF and its variants showed higher prediction accuracy as compared to other methods. We found that Naive Baye’s classifier outperforms other classifiers and achieved the highest AUC (0.72; p-value = 2.3 × 10−7) with five features: Stats_min, Wavelet_HLL_rlgl_lowGrayLevelRunEmphasis, Wavelet_HHL_stats_median, Wavelet_HLL_stats_skewness, and Wavelet_HLH_glcm_clusShade. Conclusion Histological subtypes can influence the choice of a treatment/therapy for lung cancer patients. We observed that radiomic features show significant association with the lung tumor histology. Moreover, radiomics-based multivariate classifiers were independently validated for the prediction of histological subtypes. Despite achieving lower than optimal prediction accuracy (AUC 0.72), our analysis highlights the impressive potential of non-invasive and cost-effective radiomics for precision medicine. Further research in this direction could lead us to optimal performance and therefore to clinical applicability, which could enhance the efficiency and efficacy of cancer care.
Collapse
Affiliation(s)
- Weimiao Wu
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Chintan Parmar
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Research Institute GROW, Maastricht University, Maastricht, Netherlands
| | - Patrick Grossmann
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - John Quackenbush
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Philippe Lambin
- Research Institute GROW, Maastricht University , Maastricht , Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center , Nijmegen , Netherlands
| | - Raymond Mak
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| | - Hugo J W L Aerts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
336
|
Rao A, Manyam G, Rao G, Jain R. Integrative Analysis of mRNA, microRNA, and Protein Correlates of Relative Cerebral Blood Volume Values in GBM Reveals the Role for Modulators of Angiogenesis and Tumor Proliferation. Cancer Inform 2016; 15:29-33. [PMID: 27053917 PMCID: PMC4814129 DOI: 10.4137/cin.s33014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/29/2016] [Accepted: 12/07/2015] [Indexed: 12/12/2022] Open
Abstract
Dynamic susceptibility contrast-enhanced magnetic resonance imaging is routinely used to provide hemodynamic assessment of brain tumors as a diagnostic as well as a prognostic tool. Recently, it was shown that the relative cerebral blood volume (rCBV), obtained from the contrast-enhancing as well as -nonenhancing portion of glioblastoma (GBM), is strongly associated with overall survival. In this study, we aim to characterize the genomic correlates (microRNA, messenger RNA, and protein) of this vascular parameter. This study aims to provide a comprehensive radiogenomic and radioproteomic characterization of the hemodynamic phenotype of GBM using publicly available imaging and genomic data from the Cancer Genome Atlas GBM cohort. Based on this analysis, we identified pathways associated with angiogenesis and tumor proliferation underlying this hemodynamic parameter in GBM.
Collapse
Affiliation(s)
- Arvind Rao
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganiraju Manyam
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganesh Rao
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rajan Jain
- Department of Radiology, NY University School of Medicine, New York, NY, USA
| |
Collapse
|
337
|
Wei K, Su H, Zhou G, Zhang R, Cai P, Fan Y, Xie C, Fei B, Zhang Z. Potential Application of Radiomics for Differentiating Solitary Pulmonary Nodules. ACTA ACUST UNITED AC 2016; 5. [PMID: 28261535 PMCID: PMC5336142 DOI: 10.4172/2167-7964.1000218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A solitary pulmonary nodule is defined as radiographic lesion with diameters no more than 3 cm and completely surrounded by normal lung tissue. It is commonly encountered in clinical practice and its diagnosis is a big challenge. Medical imaging, as a non-invasive approach, plays a crucial role in the diagnosis of solitary pulmonary nodules since the potential morbidity of surgery and the limits of biopsy. Advanced hardware, image acquisition and analysis technologies have led to the utilization of imaging towards quantitative imaging. With the aim of mining more useful information from image data, radiomics with high-throughput extraction can play a useful role. This article is to introduce the current state of radiomics studies and describe the general procedures. Another objective of this paper is to discover the feasibility and potential of radiomics methods on differentiating solitary pulmonary nodules and to look into the future direction of radiomics in this area.
Collapse
Affiliation(s)
- Kaikai Wei
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Huifang Su
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Guofeng Zhou
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Rong Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Peiqiang Cai
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Yi Fan
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Chuanmiao Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Baowei Fei
- Department of Radiology and Imaging Sciences, Emory University School of Medicine; Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, USA
| | - Zhenfeng Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| |
Collapse
|
338
|
Li M, Fu S, Zhu Y, Liu Z, Chen S, Lu L, Liang C. Computed tomography texture analysis to facilitate therapeutic decision making in hepatocellular carcinoma. Oncotarget 2016; 7:13248-13259. [PMID: 26910890 PMCID: PMC4914356 DOI: 10.18632/oncotarget.7467] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/27/2016] [Indexed: 02/06/2023] Open
Abstract
This study explored the potential of computed tomography (CT) textural feature analysis for the stratification of single large hepatocellular carcinomas (HCCs) > 5 cm, and the subsequent determination of patient suitability for liver resection (LR) or transcatheter arterial chemoembolization (TACE). Wavelet decomposition was performed on portal-phase CT images with three bandwidth responses (filter 0, 1.0, and 1.5). Nine textural features of each filter were extracted from regions of interest. Wavelet-2-H (filter 1.0) in LR and wavelet-2-V (filter 0 and 1.0) in TACE were related to survival. Subsequently, LR and TACE patients were divided based on the wavelet-2-H and wavelet-2-V median at filter 1.0 into two subgroups (+ or -). LR+ patients showed the best survival, followed by LR-, TACE+, and TACE-. We estimated that LR+ patients treated using TACE would exhibit a survival similar to TACE- patients and worse than TACE+ patients, with a severe compromise in overall survival. LR was recommended for TACE- patients, whereas TACE was preferred for LR- and TACE+ patients. Independent of tumor size, CT textural features showed positive and negative correlations with survival after LR and TACE, respectively. Although further validation is needed, texture analysis demonstrated the feasibility of using HCC patient stratification for determining the suitability of LR vs. TACE.
Collapse
Affiliation(s)
- Meng Li
- Southern Medical University, Guangzhou, China
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Sirui Fu
- Department of Interventional Oncology, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yanjie Zhu
- Shenzhen Institutes of Advanced Technology, Shenzhen, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shuting Chen
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ligong Lu
- Department of Interventional Oncology, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Changhong Liang
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
339
|
Benson M. Clinical implications of omics and systems medicine: focus on predictive and individualized treatment. J Intern Med 2016; 279:229-40. [PMID: 26891944 DOI: 10.1111/joim.12412] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Many patients with common diseases do not respond to treatment. This is a key challenge to modern health care, which causes both suffering and enormous costs. One important reason for the lack of treatment response is that common diseases are associated with altered interactions between thousands of genes, in combinations that differ between subgroups of patients who do or do not respond to a given treatment. Such subgroups, or even distinct disease entities, have been described recently in asthma, diabetes, autoimmune diseases and cancer. High-throughput techniques (omics) allow identification and characterization of such subgroups or entities. This may have important clinical implications, such as identification of diagnostic markers for individualized medicine, as well as new therapeutic targets for patients who do not respond to existing drugs. For example, whole-genome sequencing may be applied to more accurately guide treatment of neurodevelopmental diseases, or to identify drugs specifically targeting mutated genes in cancer. A study published in 2015 showed that 28% of hepatocellular carcinomas contained mutated genes that potentially could be targeted by drugs already approved by the US Food and Drug Administration. A translational study, which is described in detail, showed how combined omics, computational, functional and clinical studies could identify and validate a novel diagnostic and therapeutic candidate gene in allergy. Another important clinical implication is the identification of potential diagnostic markers and therapeutic targets for predictive and preventative medicine. By combining computational and experimental methods, early disease regulators may be identified and potentially used to predict and treat disease before it becomes symptomatic. Systems medicine is an emerging discipline, which may contribute to such developments through combining omics with computational, functional and clinical studies. The aims of this review are to provide a brief introduction to systems medicine and discuss how it may contribute to the clinical implementation of individualized treatment, using clinically relevant examples.
Collapse
Affiliation(s)
- M Benson
- Centre for Individualized Medicine, Department of Pediatrics, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
340
|
Bai HX, Lee AM, Yang L, Zhang P, Davatzikos C, Maris JM, Diskin SJ. Imaging genomics in cancer research: limitations and promises. Br J Radiol 2016; 89:20151030. [PMID: 26864054 DOI: 10.1259/bjr.20151030] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recently, radiogenomics or imaging genomics has emerged as a novel high-throughput method of associating imaging features with genomic data. Radiogenomics has the potential to provide comprehensive intratumour, intertumour and peritumour information non-invasively. This review article summarizes the current state of radiogenomic research in tumour characterization, discusses some of its limitations and promises and projects its future directions. Semi-radiogenomic studies that relate specific gene expressions to imaging features will also be briefly reviewed.
Collapse
Affiliation(s)
- Harrison X Bai
- 1 Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley M Lee
- 1 Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Li Yang
- 2 Department of Neurology, The Second Xiangya Hospital, Changsha, Hunan, China
| | - Paul Zhang
- 3 Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- 1 Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - John M Maris
- 4 Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,5 Abramson Family Cancer Research Institute, PerelmanSchool of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,6 Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sharon J Diskin
- 4 Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,5 Abramson Family Cancer Research Institute, PerelmanSchool of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,6 Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
341
|
Calcagno C, Mulder WJM, Nahrendorf M, Fayad ZA. Systems Biology and Noninvasive Imaging of Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 36:e1-8. [PMID: 26819466 PMCID: PMC4861402 DOI: 10.1161/atvbaha.115.306350] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Claudia Calcagno
- From the Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.C., W.J.M.M., Z.A.F.); Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands (W.J.M.M.); and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (M.N.).
| | - Willem J M Mulder
- From the Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.C., W.J.M.M., Z.A.F.); Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands (W.J.M.M.); and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (M.N.)
| | - Matthias Nahrendorf
- From the Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.C., W.J.M.M., Z.A.F.); Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands (W.J.M.M.); and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (M.N.)
| | - Zahi A Fayad
- From the Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.C., W.J.M.M., Z.A.F.); Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands (W.J.M.M.); and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA (M.N.)
| |
Collapse
|
342
|
Blackledge MD, Collins DJ, Koh DM, Leach MO. Rapid development of image analysis research tools: Bridging the gap between researcher and clinician with pyOsiriX. Comput Biol Med 2016; 69:203-12. [PMID: 26773941 PMCID: PMC4761020 DOI: 10.1016/j.compbiomed.2015.12.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/07/2015] [Accepted: 12/03/2015] [Indexed: 01/08/2023]
Abstract
We present pyOsiriX, a plugin built for the already popular dicom viewer OsiriX that provides users the ability to extend the functionality of OsiriX through simple Python scripts. This approach allows users to integrate the many cutting-edge scientific/image-processing libraries created for Python into a powerful DICOM visualisation package that is intuitive to use and already familiar to many clinical researchers. Using pyOsiriX we hope to bridge the apparent gap between basic imaging scientists and clinical practice in a research setting and thus accelerate the development of advanced clinical image processing. We provide arguments for the use of Python as a robust scripting language for incorporation into larger software solutions, outline the structure of pyOsiriX and how it may be used to extend the functionality of OsiriX, and we provide three case studies that exemplify its utility. For our first case study we use pyOsiriX to provide a tool for smooth histogram display of voxel values within a user-defined region of interest (ROI) in OsiriX. We used a kernel density estimation (KDE) method available in Python using the scikit-learn library, where the total number of lines of Python code required to generate this tool was 22. Our second example presents a scheme for segmentation of the skeleton from CT datasets. We have demonstrated that good segmentation can be achieved for two example CT studies by using a combination of Python libraries including scikit-learn, scikit-image, SimpleITK and matplotlib. Furthermore, this segmentation method was incorporated into an automatic analysis of quantitative PET-CT in a patient with bone metastases from primary prostate cancer. This enabled repeatable statistical evaluation of PET uptake values for each lesion, before and after treatment, providing estaimes maximum and median standardised uptake values (SUVmax and SUVmed respectively). Following treatment we observed a reduction in lesion volume, SUVmax and SUVmed for all lesions, in agreement with a reduction in concurrent measures of serum prostate-specific antigen (PSA).
Collapse
Affiliation(s)
- Matthew D Blackledge
- CR-UK Cancer Imaging Centre, Radiotherapy and Imaging Division, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom.
| | - David J Collins
- CR-UK Cancer Imaging Centre, Radiotherapy and Imaging Division, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Dow-Mu Koh
- CR-UK Cancer Imaging Centre, Radiotherapy and Imaging Division, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Martin O Leach
- CR-UK Cancer Imaging Centre, Radiotherapy and Imaging Division, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
343
|
Abstract
In the past decade, the field of medical image analysis has grown exponentially, with an increased number of pattern recognition tools and an increase in data set sizes. These advances have facilitated the development of processes for high-throughput extraction of quantitative features that result in the conversion of images into mineable data and the subsequent analysis of these data for decision support; this practice is termed radiomics. This is in contrast to the traditional practice of treating medical images as pictures intended solely for visual interpretation. Radiomic data contain first-, second-, and higher-order statistics. These data are combined with other patient data and are mined with sophisticated bioinformatics tools to develop models that may potentially improve diagnostic, prognostic, and predictive accuracy. Because radiomics analyses are intended to be conducted with standard of care images, it is conceivable that conversion of digital images to mineable data will eventually become routine practice. This report describes the process of radiomics, its challenges, and its potential power to facilitate better clinical decision making, particularly in the care of patients with cancer.
Collapse
Affiliation(s)
- Robert J. Gillies
- From the Department of Cancer Imaging, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612 (R.J.G.); Department of Radiology, University of Washington, Seattle, Wash (P.E.K.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York NY 10065 (H.H.)
| | - Paul E. Kinahan
- From the Department of Cancer Imaging, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612 (R.J.G.); Department of Radiology, University of Washington, Seattle, Wash (P.E.K.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York NY 10065 (H.H.)
| | - Hedvig Hricak
- From the Department of Cancer Imaging, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612 (R.J.G.); Department of Radiology, University of Washington, Seattle, Wash (P.E.K.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York NY 10065 (H.H.)
| |
Collapse
|
344
|
Tan M, Li Z, Qiu Y, McMeekin SD, Thai TC, Ding K, Moore KN, Liu H, Zheng B. A New Approach to Evaluate Drug Treatment Response of Ovarian Cancer Patients Based on Deformable Image Registration. IEEE TRANSACTIONS ON MEDICAL IMAGING 2016; 35:316-325. [PMID: 26336119 PMCID: PMC5161344 DOI: 10.1109/tmi.2015.2473823] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Although Response Evaluation Criteria in Solid Tumors (RECIST) is the current clinical guideline to assess size change of solid tumors after therapeutic treatment, it has a relatively lower association to the clinical outcome of progression free survival (PFS) of the patients. In this paper, we presented a new approach to assess responses of ovarian cancer patients to new chemotherapy drugs in clinical trials. We first developed and applied a multi-resolution B-spline based deformable image registration method to register two sets of computed tomography (CT) image data acquired pre- and post-treatment. The B-spline difference maps generated from the co-registered CT images highlight the regions related to the volumetric growth or shrinkage of the metastatic tumors, and density changes related to variation of necrosis inside the solid tumors. Using a testing dataset involving 19 ovarian cancer patients, we compared patients' response to the treatment using the new image registration method and RECIST guideline. The results demonstrated that using the image registration method yielded higher association with the six-month PFS outcomes of the patients than using RECIST. The image registration results also provided a solid foundation of developing new computerized quantitative image feature analysis schemes in the future studies.
Collapse
Affiliation(s)
| | - Zheng Li
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019 USA
| | - Yuchen Qiu
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019 USA
| | - Scott D. McMeekin
- Health Science Center of University of Oklahoma, Oklahoma City, OK 73104 USA
| | - Theresa C. Thai
- Health Science Center of University of Oklahoma, Oklahoma City, OK 73104 USA
| | - Kai Ding
- Health Science Center of University of Oklahoma, Oklahoma City, OK 73104 USA
| | - Kathleen N. Moore
- Health Science Center of University of Oklahoma, Oklahoma City, OK 73104 USA
| | - Hong Liu
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019 USA
| | - Bin Zheng
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019 USA
| |
Collapse
|
345
|
Napel S, Giger M. Special Section Guest Editorial:Radiomics and Imaging Genomics: Quantitative Imaging for Precision Medicine. J Med Imaging (Bellingham) 2015; 2:041001. [PMID: 26839908 DOI: 10.1117/1.jmi.2.4.041001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Sandy Napel
- Stanford University School of Medicine , Radiology Department , 318 Campus Drive #S323 , Stanford, California 94305-5014
| | - Maryellen Giger
- The University of Chicago , Radiology Department , 5841 S. Maryland Avenue , Chicago, Illinois 60637-1447
| |
Collapse
|
346
|
Renzulli M, Brocchi S, Cucchetti A, Mazzotti F, Mosconi C, Sportoletti C, Brandi G, Pinna AD, Golfieri R. Can Current Preoperative Imaging Be Used to Detect Microvascular Invasion of Hepatocellular Carcinoma? Radiology 2015; 279:432-42. [PMID: 26653683 DOI: 10.1148/radiol.2015150998] [Citation(s) in RCA: 283] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE To determine the accuracy of imaging features, such as tumor dimension, multinodularity, nonsmooth tumor margins, peritumoral enhancement, and radiogenomic algorithm based on the association between imaging features (internal arteries and hypoattenuating halos) and gene expression that the authors called two-trait predictor of venous invasion (TTPVI), in the prediction of microvascular invasion (MVI) in hepatocellular carcinoma (HCC). MATERIALS AND METHODS This single-center retrospective study was approved by the institutional review board, and the requirement for informed consent was waived. One hundred twenty-five patients (median age, 63 years; interquartile range, 53-71 years) with a diagnosis of HCC and indications for hepatic resection were included. Two observers independently reviewed radiologic images to evaluate the following features for MVI: maximum diameter, number of lesions, tumor margins, TTPVI, and peritumoral enhancement. Interobserver agreement was checked, and diagnostic accuracy of radiologic features was investigated. RESULTS The total number of HCC nodules was 140. Large tumor size, nonsmooth tumor margins, TTPVI, and peritumoral enhancement were significantly related to the presence of MVI (P < .05 in all cases and for both observers). Multinodularity was not significantly related (P = .158). Moreover, the diagnostic accuracy of the three "worrisome" radiologic features (nonsmooth tumor margins, peritumoral enhancement, and TTPVI) was associated with tumor size: The negative predictive value of the absence of worrisome features decreased from 0.84 for observer 1 and 0.91 for observer 2 for tumors smaller than 2 cm to 0.56 and 0.71, respectively, for tumors larger than 5 cm, whereas the presence of all three worrisome features returned to a positive predictive value of 0.95 for observer 1 and 0.96 for observer 2 independent of tumor size, with no significant interobserver differences (P > .10). CONCLUSION "Worrisome" imaging features, such as tumor dimension, nonsmooth tumor margins, peritumoral enhancement, and TTPVI, have high accuracy in the prediction of MVI in HCC.
Collapse
Affiliation(s)
- Matteo Renzulli
- From the Radiology Unit, Department of Diagnostic Medicine and Prevention (M.R., S.B., C.M., C.S., R.G.), Department of Medical and Surgical Sciences (A.C., F.M., A.D.P.), and Department of Specialized, Experimental and Diagnostic Medicine (G.B.), S. Orsola-Malpighi Hospital, University of Bologna, Via Albertoni 15, 40138 Bologna, Italy
| | - Stefano Brocchi
- From the Radiology Unit, Department of Diagnostic Medicine and Prevention (M.R., S.B., C.M., C.S., R.G.), Department of Medical and Surgical Sciences (A.C., F.M., A.D.P.), and Department of Specialized, Experimental and Diagnostic Medicine (G.B.), S. Orsola-Malpighi Hospital, University of Bologna, Via Albertoni 15, 40138 Bologna, Italy
| | - Alessandro Cucchetti
- From the Radiology Unit, Department of Diagnostic Medicine and Prevention (M.R., S.B., C.M., C.S., R.G.), Department of Medical and Surgical Sciences (A.C., F.M., A.D.P.), and Department of Specialized, Experimental and Diagnostic Medicine (G.B.), S. Orsola-Malpighi Hospital, University of Bologna, Via Albertoni 15, 40138 Bologna, Italy
| | - Federico Mazzotti
- From the Radiology Unit, Department of Diagnostic Medicine and Prevention (M.R., S.B., C.M., C.S., R.G.), Department of Medical and Surgical Sciences (A.C., F.M., A.D.P.), and Department of Specialized, Experimental and Diagnostic Medicine (G.B.), S. Orsola-Malpighi Hospital, University of Bologna, Via Albertoni 15, 40138 Bologna, Italy
| | - Cristina Mosconi
- From the Radiology Unit, Department of Diagnostic Medicine and Prevention (M.R., S.B., C.M., C.S., R.G.), Department of Medical and Surgical Sciences (A.C., F.M., A.D.P.), and Department of Specialized, Experimental and Diagnostic Medicine (G.B.), S. Orsola-Malpighi Hospital, University of Bologna, Via Albertoni 15, 40138 Bologna, Italy
| | - Camilla Sportoletti
- From the Radiology Unit, Department of Diagnostic Medicine and Prevention (M.R., S.B., C.M., C.S., R.G.), Department of Medical and Surgical Sciences (A.C., F.M., A.D.P.), and Department of Specialized, Experimental and Diagnostic Medicine (G.B.), S. Orsola-Malpighi Hospital, University of Bologna, Via Albertoni 15, 40138 Bologna, Italy
| | - Giovanni Brandi
- From the Radiology Unit, Department of Diagnostic Medicine and Prevention (M.R., S.B., C.M., C.S., R.G.), Department of Medical and Surgical Sciences (A.C., F.M., A.D.P.), and Department of Specialized, Experimental and Diagnostic Medicine (G.B.), S. Orsola-Malpighi Hospital, University of Bologna, Via Albertoni 15, 40138 Bologna, Italy
| | - Antonio Daniele Pinna
- From the Radiology Unit, Department of Diagnostic Medicine and Prevention (M.R., S.B., C.M., C.S., R.G.), Department of Medical and Surgical Sciences (A.C., F.M., A.D.P.), and Department of Specialized, Experimental and Diagnostic Medicine (G.B.), S. Orsola-Malpighi Hospital, University of Bologna, Via Albertoni 15, 40138 Bologna, Italy
| | - Rita Golfieri
- From the Radiology Unit, Department of Diagnostic Medicine and Prevention (M.R., S.B., C.M., C.S., R.G.), Department of Medical and Surgical Sciences (A.C., F.M., A.D.P.), and Department of Specialized, Experimental and Diagnostic Medicine (G.B.), S. Orsola-Malpighi Hospital, University of Bologna, Via Albertoni 15, 40138 Bologna, Italy
| |
Collapse
|
347
|
Parmar C, Grossmann P, Rietveld D, Rietbergen MM, Lambin P, Aerts HJWL. Radiomic Machine-Learning Classifiers for Prognostic Biomarkers of Head and Neck Cancer. Front Oncol 2015; 5:272. [PMID: 26697407 PMCID: PMC4668290 DOI: 10.3389/fonc.2015.00272] [Citation(s) in RCA: 259] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/20/2015] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION "Radiomics" extracts and mines a large number of medical imaging features in a non-invasive and cost-effective way. The underlying assumption of radiomics is that these imaging features quantify phenotypic characteristics of an entire tumor. In order to enhance applicability of radiomics in clinical oncology, highly accurate and reliable machine-learning approaches are required. In this radiomic study, 13 feature selection methods and 11 machine-learning classification methods were evaluated in terms of their performance and stability for predicting overall survival in head and neck cancer patients. METHODS Two independent head and neck cancer cohorts were investigated. Training cohort HN1 consisted of 101 head and neck cancer patients. Cohort HN2 (n = 95) was used for validation. A total of 440 radiomic features were extracted from the segmented tumor regions in CT images. Feature selection and classification methods were compared using an unbiased evaluation framework. RESULTS We observed that the three feature selection methods minimum redundancy maximum relevance (AUC = 0.69, Stability = 0.66), mutual information feature selection (AUC = 0.66, Stability = 0.69), and conditional infomax feature extraction (AUC = 0.68, Stability = 0.7) had high prognostic performance and stability. The three classifiers BY (AUC = 0.67, RSD = 11.28), RF (AUC = 0.61, RSD = 7.36), and NN (AUC = 0.62, RSD = 10.52) also showed high prognostic performance and stability. Analysis investigating performance variability indicated that the choice of classification method is the major factor driving the performance variation (29.02% of total variance). CONCLUSION Our study identified prognostic and reliable machine-learning methods for the prediction of overall survival of head and neck cancer patients. Identification of optimal machine-learning methods for radiomics-based prognostic analyses could broaden the scope of radiomics in precision oncology and cancer care.
Collapse
Affiliation(s)
- Chintan Parmar
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Radiation Oncology (MAASTRO), Research Institute GROW, Maastricht University , Maastricht , Netherlands
| | - Patrick Grossmann
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute , Boston, MA , USA
| | - Derek Rietveld
- Department of Radiation Oncology, VU University Medical Center , Amsterdam , Netherlands
| | - Michelle M Rietbergen
- Department of Otolaryngology/Head and Neck Surgery, VU University Medical Center , Amsterdam , Netherlands
| | - Philippe Lambin
- Radiation Oncology (MAASTRO), Research Institute GROW, Maastricht University , Maastricht , Netherlands
| | - Hugo J W L Aerts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute , Boston, MA , USA
| |
Collapse
|
348
|
Tam AL, Lim HJ, Wistuba II, Tamrazi A, Kuo MD, Ziv E, Wong S, Shih AJ, Webster RJ, Fischer GS, Nagrath S, Davis SE, White SB, Ahrar K. Image-Guided Biopsy in the Era of Personalized Cancer Care: Proceedings from the Society of Interventional Radiology Research Consensus Panel. J Vasc Interv Radiol 2015; 27:8-19. [PMID: 26626860 DOI: 10.1016/j.jvir.2015.10.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 10/23/2015] [Accepted: 10/23/2015] [Indexed: 02/07/2023] Open
Affiliation(s)
- Alda L Tam
- Departments of Interventional Radiology, Houston, Texas.
| | - Howard J Lim
- Division of Medical Oncology, University of British Columbia, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | | | - Anobel Tamrazi
- Division of Vascular and Interventional Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael D Kuo
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Etay Ziv
- Departments of Interventional Radiology and Computational Biology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Stephen Wong
- Department of Systems Medicine & Bioengineering, Houston Methodist Research Institute, Houston, Texas
| | - Albert J Shih
- Departments of Mechanical and Biomechanical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Robert J Webster
- Department of Mechanical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Gregory S Fischer
- Automation and Interventional Medicine Robotics Lab, Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Sunitha Nagrath
- Chemical and Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Suzanne E Davis
- Division of Cancer Medicine, Research Planning and Development, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Sarah B White
- Department of Systems Medicine & Bioengineering, Houston Methodist Research Institute, Houston, Texas; Departments of Radiology, Neuroscience, Pathology & Laboratory Medicine, Weill Cornell Medical College of Cornell University, New York, New York; Division of Vascular and Interventional Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kamran Ahrar
- Departments of Interventional Radiology, Houston, Texas
| |
Collapse
|
349
|
Jamshidi N, Jonasch E, Zapala M, Korn RL, Brooks JD, Ljungberg B, Kuo MD. The radiogenomic risk score stratifies outcomes in a renal cell cancer phase 2 clinical trial. Eur Radiol 2015; 26:2798-807. [PMID: 26560727 DOI: 10.1007/s00330-015-4082-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 09/30/2015] [Accepted: 10/23/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVES To characterize a radiogenomic risk score (RRS), a previously defined biomarker, and to evaluate its potential for stratifying radiological progression-free survival (rPFS) in patients with metastatic renal cell carcinoma (mRCC) undergoing pre-surgical treatment with bevacizumab. METHODOLOGY In this IRB-approved study, prospective imaging analysis of the RRS was performed on phase II clinical trial data of mRCC patients (n = 41) evaluating whether patient stratification according to the RRS resulted in groups more or less likely to have a rPFS to pre-surgical bevacizumab prior to cytoreductive nephrectomy. Survival times of RRS subgroups were analyzed using Kaplan-Meier survival analysis. RESULTS The RRS is enriched in diverse molecular processes including drug response, stress response, protein kinase regulation, and signal transduction pathways (P < 0.05). The RRS successfully stratified rPFS to bevacizumab based on pre-treatment computed tomography imaging with a median progression-free survival of 6 versus >25 months (P = 0.005) and overall survival of 25 versus >37 months in the high and low RRS groups (P = 0.03), respectively. Conventional prognostic predictors including the Motzer and Heng criteria were not predictive in this cohort (P > 0.05). CONCLUSIONS The RRS stratifies rPFS to bevacizumab in patients from a phase II clinical trial with mRCC undergoing cytoreductive nephrectomy and pre-surgical bevacizumab. KEY POINTS • The RRS SOMA stratifies patient outcomes in a phase II clinical trial. • RRS stratifies subjects into prognostic groups in a discrete or continuous fashion. • RRS is biologically enriched in diverse processes including drug response programs.
Collapse
Affiliation(s)
- Neema Jamshidi
- Department of Radiological Sciences, University of California-Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Matthew Zapala
- Department of Radiological Sciences, University of California-Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
- Department of Radiology, University of California-San Diego, San Diego, CA, USA
| | | | - James D Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Borje Ljungberg
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umea Hospital, Umea, Sweden
| | - Michael D Kuo
- Department of Radiological Sciences, University of California-Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
350
|
Herold CJ, Lewin JS, Wibmer AG, Thrall JH, Krestin GP, Dixon AK, Schoenberg SO, Geckle RJ, Muellner A, Hricak H. Imaging in the Age of Precision Medicine: Summary of the Proceedings of the 10th Biannual Symposium of the International Society for Strategic Studies in Radiology. Radiology 2015; 279:226-38. [PMID: 26465058 DOI: 10.1148/radiol.2015150709] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
During the past decade, with its breakthroughs in systems biology, precision medicine (PM) has emerged as a novel health-care paradigm. Challenging reductionism and broad-based approaches in medicine, PM is an approach for disease treatment and prevention that takes into account individual variability in genes, environment, and lifestyle. It involves integrating information from multiple sources in a holistic manner to achieve a definitive diagnosis, focused treatment, and adequate response assessment. Biomedical imaging and imaging-guided interventions, which provide multiparametric morphologic and functional information and enable focused, minimally invasive treatments, are key elements in the infrastructure needed for PM. The emerging discipline of radiogenomics, which links genotypic information to phenotypic disease manifestations at imaging, should also greatly contribute to patient-tailored care. Because of the growing volume and complexity of imaging data, decision-support algorithms will be required to help physicians apply the most essential patient data for optimal management. These innovations will challenge traditional concepts of health care and business models. Reimbursement policies and quality assurance measures will have to be reconsidered and adapted. In their 10th biannual symposium, which was held in August 2013, the members of the International Society for Strategic Studies in Radiology discussed the opportunities and challenges arising for the imaging community with the transition to PM. This article summarizes the discussions and central messages of the symposium.
Collapse
Affiliation(s)
- Christian J Herold
- From the Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria (C.J.H., A.G.W.); Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (J.S.L., R.J.G.); Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (J.H.T.); Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands (G.P.K.); Department of Radiology, University of Cambridge, Cambridge, England (A.K.D.); Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (S.O.S.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C-278, New York, NY 10065 (A.M., H.H.)
| | - Jonathan S Lewin
- From the Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria (C.J.H., A.G.W.); Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (J.S.L., R.J.G.); Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (J.H.T.); Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands (G.P.K.); Department of Radiology, University of Cambridge, Cambridge, England (A.K.D.); Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (S.O.S.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C-278, New York, NY 10065 (A.M., H.H.)
| | - Andreas G Wibmer
- From the Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria (C.J.H., A.G.W.); Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (J.S.L., R.J.G.); Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (J.H.T.); Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands (G.P.K.); Department of Radiology, University of Cambridge, Cambridge, England (A.K.D.); Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (S.O.S.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C-278, New York, NY 10065 (A.M., H.H.)
| | - James H Thrall
- From the Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria (C.J.H., A.G.W.); Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (J.S.L., R.J.G.); Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (J.H.T.); Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands (G.P.K.); Department of Radiology, University of Cambridge, Cambridge, England (A.K.D.); Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (S.O.S.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C-278, New York, NY 10065 (A.M., H.H.)
| | - Gabriel P Krestin
- From the Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria (C.J.H., A.G.W.); Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (J.S.L., R.J.G.); Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (J.H.T.); Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands (G.P.K.); Department of Radiology, University of Cambridge, Cambridge, England (A.K.D.); Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (S.O.S.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C-278, New York, NY 10065 (A.M., H.H.)
| | - Adrian K Dixon
- From the Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria (C.J.H., A.G.W.); Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (J.S.L., R.J.G.); Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (J.H.T.); Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands (G.P.K.); Department of Radiology, University of Cambridge, Cambridge, England (A.K.D.); Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (S.O.S.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C-278, New York, NY 10065 (A.M., H.H.)
| | - Stefan O Schoenberg
- From the Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria (C.J.H., A.G.W.); Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (J.S.L., R.J.G.); Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (J.H.T.); Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands (G.P.K.); Department of Radiology, University of Cambridge, Cambridge, England (A.K.D.); Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (S.O.S.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C-278, New York, NY 10065 (A.M., H.H.)
| | - Rena J Geckle
- From the Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria (C.J.H., A.G.W.); Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (J.S.L., R.J.G.); Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (J.H.T.); Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands (G.P.K.); Department of Radiology, University of Cambridge, Cambridge, England (A.K.D.); Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (S.O.S.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C-278, New York, NY 10065 (A.M., H.H.)
| | - Ada Muellner
- From the Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria (C.J.H., A.G.W.); Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (J.S.L., R.J.G.); Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (J.H.T.); Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands (G.P.K.); Department of Radiology, University of Cambridge, Cambridge, England (A.K.D.); Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (S.O.S.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C-278, New York, NY 10065 (A.M., H.H.)
| | - Hedvig Hricak
- From the Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria (C.J.H., A.G.W.); Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (J.S.L., R.J.G.); Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass (J.H.T.); Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands (G.P.K.); Department of Radiology, University of Cambridge, Cambridge, England (A.K.D.); Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (S.O.S.); and Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C-278, New York, NY 10065 (A.M., H.H.)
| |
Collapse
|