201
|
Ma Y, Feng W, Wu Z, Liu M, Zhang F, Liang Z, Cui C, Huang J, Li X, Guo X. Intra-tumoural heterogeneity characterization through texture and colour analysis for differentiation of non-small cell lung carcinoma subtypes. Phys Med Biol 2018; 63:165018. [PMID: 30051884 DOI: 10.1088/1361-6560/aad648] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Radiomics has shown potential in disease diagnosis, but its feasibility for non-small cell lung carcinoma (NSCLC) subtype classification is unclear. This study aims to explore the diagnosis value of texture and colour features from positron emission tomography computed tomography (PET-CT) images in differentiation of NSCLC subtypes: adenocarcinoma (ADC) and squamous cell carcinoma (SqCC). Two patient cohorts were retrospectively collected into a dataset of 341 18F-labeled 2-deoxy-2fluoro-d-glucose ([18F] FDG) PET-CT images of NSCLC tumours (125 ADC, 174 SqCC, and 42 cases with unknown subtype). Quantification of texture and colour features was performed using freehand regions of interest. The relation between extracted features and commonly used parameters such as age, gender, tumour size, and standard uptake value (SUVmax) was explored. To classify NSCLC subtypes, support vector machine algorithm was applied on these features and the classification performance was evaluated by receiver operating characteristic curve analysis. There was a significant difference between ADC and SqCC subtypes in texture and colour features (P < 0.05); this showed that imaging features were significantly correlated to both SUVmax and tumour diameter (P < 0.05). When evaluating classification performance, features combining texture and colour showed an AUC of 0.89 (95% CI, 0.78-1.00), colour features showed an AUC of 0.85 (95% CI, 0.71-0.99), and texture features showed an AUC of 0.68 (95% CI, 0.48-0.88). DeLong's test showed that AUC was higher for features combining texture and colour than that for texture features only (P = 0.010), but not significantly different from that for colour features only (P = 0.328). HSV colour features showed a similar performance to RGB colour features (P = 0.473). The colour features are promising in the refinement of NSCLC subtype differentiation, and features combining texture and colour of PET-CT images could result in better classification performance.
Collapse
Affiliation(s)
- Yuan Ma
- School of Public Health, Capital Medical University, Beijing, People's Republic of China. Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
202
|
van Helden EJ, Vacher YJL, van Wieringen WN, van Velden FHP, Verheul HMW, Hoekstra OS, Boellaard R, Menke-van der Houven van Oordt CW. Radiomics analysis of pre-treatment [ 18F]FDG PET/CT for patients with metastatic colorectal cancer undergoing palliative systemic treatment. Eur J Nucl Med Mol Imaging 2018; 45:2307-2317. [PMID: 30094460 PMCID: PMC6208805 DOI: 10.1007/s00259-018-4100-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/17/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND The aim of this study was to assess radiomics features on pre-treatment [18F]FDG positron emission tomography (PET) as potential biomarkers for response and survival in patients with metastatic colorectal cancer (mCRC). METHODS Patients with mCRC underwent [18F]FDG PET/computed tomography (CT) prior to first- or third-line palliative systemic treatment. Tumour lesions were semiautomatically delineated and standard uptake value (SUV), metabolically active tumour volume (MATV), total lesion glycolysis (TLG), entropy, area under the curve of the cumulative SUV-volume histogram (AUC-CSH), compactness and sphericity were obtained. RESULTS Lesions of 47 patients receiving third-line systemic treatment had higher SUVmax, SUVpeak, SUVmean, MATV and TLG, and lower AUC-CSH, compactness and sphericity compared to 52 patients receiving first-line systemic treatment. Therefore, first- and third-line groups were evaluated separately. In the first-line group, anatomical changes on CT correlated negatively with TLG (ρ = 0.31) and MATV (ρ = 0.36), and positively with compactness (ρ = -0.27) and sphericity (ρ = -0.27). Patients without benefit had higher mean entropy (p = 0.021). Progression-free survival (PFS) and overall survival (OS) were worse with a decreased mean AUC [hazard ratio (HR) 0.86, HR 0.77] and increase in mean MATV (HR 1.15, HR 1.22), sum MATV (HR 1.14, HR 1.19), mean TLG (HR 1.16, HR 1.22) and sum TLG (HT1.12, HR1.18). In the third-line group, AUC-CSH correlated negatively with anatomical change (ρ = 0.21). PFS and OS were worse with an increased mean MATV (HR 1.27, HR 1.68), sum MATV (HR 1.35, HR 2.04), mean TLG (HR 1.29, HR 1.52) and sum TLG (HT 1.27, HR 1.80). SUVmax and SUVpeak negatively correlated with OS (HR 1.19, HR 1.21). Cluster analysis of the 10 radiomics features demonstrated no complementary value in identifying aggressively growing lesions or patients with impaired survival. CONCLUSION We demonstrated an association between improved clinical outcome and pre-treatment low tumour volume and heterogeneity as well as high sphericity on [18F]FDG PET. Future PET imaging research should include radiomics features that incorporate tumour volume and heterogeneity when correlating PET data with clinical outcome.
Collapse
Affiliation(s)
- E J van Helden
- Cancer Center Amsterdam, Department of Medical Oncology, VUmc, Amsterdam, the Netherlands
| | - Y J L Vacher
- Cancer Center Amsterdam, Department of Medical Oncology, VUmc, Amsterdam, the Netherlands
| | - W N van Wieringen
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, the Netherlands
| | - F H P van Velden
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - H M W Verheul
- Cancer Center Amsterdam, Department of Medical Oncology, VUmc, Amsterdam, the Netherlands
| | - O S Hoekstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - R Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | | |
Collapse
|
203
|
Li Z, Yu L, Wang X, Yu H, Gao Y, Ren Y, Wang G, Zhou X. Diagnostic Performance of Mammographic Texture Analysis in the Differential Diagnosis of Benign and Malignant Breast Tumors. Clin Breast Cancer 2018; 18:e621-e627. [DOI: 10.1016/j.clbc.2017.11.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 10/29/2017] [Accepted: 11/02/2017] [Indexed: 10/18/2022]
|
204
|
Integrated texture parameter of 18F-FDG PET may be a stratification factor for the survival of nonoperative patients with locally advanced non-small-cell lung cancer. Nucl Med Commun 2018; 39:732-740. [DOI: 10.1097/mnm.0000000000000860] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
205
|
Palmisano A, Esposito A, Rancoita PMV, Di Chiara A, Passoni P, Slim N, Campolongo M, Albarello L, Fiorino C, Rosati R, Del Maschio A, De Cobelli F. Could perfusion heterogeneity at dynamic contrast-enhanced MRI be used to predict rectal cancer sensitivity to chemoradiotherapy? Clin Radiol 2018; 73:911.e1-911.e7. [PMID: 30029837 DOI: 10.1016/j.crad.2018.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 06/04/2018] [Indexed: 12/16/2022]
Abstract
AIM To evaluate whether perfusion heterogeneity of rectal cancer prior to chemoradiotherapy (CRT) using histogram analysis of dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) quantitative parameters can predict response to treatment. MATERIALS AND METHODS Twenty-one patients with histologically proven rectal adenocarcinoma were enrolled prospectively. All patients underwent 1.5 T DCE-MRI before CRT. Tumour volumes were drawn on Ktrans and Ve maps, using T2-weighted (W) images as reference, and the following first-order texture parameters of Ve and Ktrans values were extracted: 25th, 50th, 75th percentile, mean, standard deviation, skewness, and kurtosis. After CRT, patients underwent surgery and according with Rödel's tumour regression grade (TRG), they were classified as poor responders "non-GR" (TRG 0-2) and good responders "GR" (TRG 3-4). Differences between GR and non-GR in DCE-MRI first-order texture parameters were evaluated using the Mann-Whitney test, and their role in the prediction of response was investigated using receiver operating characteristic (ROC) curve analysis. RESULTS Sixteen (76%) patients were classified as GR and five (24%) were non-GR. Skewness and kurtosis of Ve was significantly higher in non-GR (4.886±1.320 and 36.402±24.486, respectively) than in GR patients (1.809±1.280, p=0.003 and 6.268±8.130, p= 0.011). Ve skewness <3.635 was able to predict GR with an area under the ROC curve (AUC) of 0.988, sensitivity 93.8%, specificity 80%, and accuracy 90.5%. Ve kurtosis <21.095 was able to predict response with an AUC of 0.963, sensitivity 93.8%, specificity 80%, and accuracy 90.5%. Other parameters were not different between groups or predictors of response. CONCLUSION Ve skewness and kurtosis seem to be promising in the prediction of response to CRT in rectal cancer patients.
Collapse
Affiliation(s)
- A Palmisano
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - A Esposito
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - P M V Rancoita
- University Centre of Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, Milan, Italy
| | - A Di Chiara
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - P Passoni
- Unit of Radiotherapy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - N Slim
- Unit of Radiotherapy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - M Campolongo
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | - L Albarello
- Department of Pathology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - C Fiorino
- Medical Physics, San Raffaele Hospital, Milan, Italy
| | - R Rosati
- Vita-Salute San Raffaele University, Milan, Italy; Department of Gastrointestinal Surgery, San Raffaele Hospital, Milan, Italy
| | - A Del Maschio
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - F De Cobelli
- Unit of Clinical Research in Radiology, Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
206
|
Abstract
Although visual assessment using the Deauville criteria is strongly recommended by guidelines for treatment response monitoring in all FDG-avid lymphoma histologies, the high rate of false-positives and concerns about interobserver variability have motivated the development of quantitative tools to facilitate objective measurement of tumor response in both routine and clinical trial settings. Imaging studies using functional quantitative measures play a significant role in profiling oncologic processes. These quantitative metrics allow for objective end points in multicenter clinical trials. However, the standardization of imaging procedures including image acquisition parameters, reconstruction and analytic measures, and validation of these methods are essential to enable an individualized treatment approach. A robust quality control program associated with the inclusion of proper scanner calibration, cross-calibration with dose calibrators and across other scanners is required for accurate quantitative measurements. In this section, we will review the technical and methodological considerations related to PET-derived quantitative metrics and the relevant published data to emphasize the potential value of these metrics in the prediction of patient prognosis in lymphoma.
Collapse
Affiliation(s)
- Lale Kostakoglu
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Stéphane Chauvie
- Department of Medical Physics, 'Santa Croce e Carle' Hospital, Cuneo, Italy
| |
Collapse
|
207
|
Ceriani L, Milan L, Martelli M, Ferreri AJM, Cascione L, Zinzani PL, Di Rocco A, Conconi A, Stathis A, Cavalli F, Bellei M, Cozens K, Porro E, Giovanella L, Johnson PW, Zucca E. Metabolic heterogeneity on baseline 18FDG-PET/CT scan is a predictor of outcome in primary mediastinal B-cell lymphoma. Blood 2018; 132:179-186. [PMID: 29720487 DOI: 10.1182/blood-2018-01-826958] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/05/2018] [Indexed: 12/19/2022] Open
Abstract
An important unmet need in the management of primary mediastinal B-cell lymphoma (PMBCL) is to identify the patients for whom first-line therapy will fail to intervene before the lymphoma becomes refractory. High heterogeneity of intratumoral 18F-fluorodeoxyglucose (18FDG) uptake distribution on positron emission tomography/computed tomography (PET/CT) scans has been suggested as a possible marker of chemoresistance in solid tumors. In the present study, we investigated the prognostic value of metabolic heterogeneity (MH) in 103 patients with PMBCL prospectively enrolled in the International Extranodal Lymphoma Study Group (IELSG) 26 study, aimed at clarifying the role of PET in this lymphoma subtype. MH was estimated using the area under curve of cumulative standardized uptake value-volume histogram (AUC-CSH) method. Progression-free survival at 5 years was 94% vs 73% in low- and high-MH groups, respectively (P = .0001). In a Cox model of progression-free survival including dichotomized MH, metabolic tumor volume, total lesion glycolysis (TLG), international prognostic index, and tumor bulk (mediastinal mass > 10 cm), as well as age as a continuous variable, only TLG (P < .001) and MH (P < .001) retained statistical significance. Using these 2 features to construct a simple prognostic model resulted in early and accurate (positive predictive value, 89%; negative predictive value, ≥90%) identification of patients at high risk for progression at a point that would allow the use of risk-adapted treatments. This may provide an important opportunity for the design of future trials aimed at helping the minority of patients who harbor chemorefractory PMBCL. The study is registered at ClinicalTrials.gov as NCT00944567.
Collapse
Affiliation(s)
- Luca Ceriani
- Nuclear Medicine and PET/CT Centre, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Lisa Milan
- Nuclear Medicine and PET/CT Centre, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Maurizio Martelli
- Department of Cellular Biotechnologies and Hematology, Sapienza University, Rome, Italy
| | - Andrés J M Ferreri
- Department of Onco-Hematology, Unit of Lymphoid Malignancies, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Pier Luigi Zinzani
- Institute of Hematology "Seràgnoli", University of Bologna, Bologna Italy
| | - Alice Di Rocco
- Department of Cellular Biotechnologies and Hematology, Sapienza University, Rome, Italy
| | | | - Anastasios Stathis
- Division of Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | | | - Monica Bellei
- Department of Diagnostic, Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Elena Porro
- Institute of Oncology Research, Bellinzona, Switzerland
| | - Luca Giovanella
- Nuclear Medicine and PET/CT Centre, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Peter W Johnson
- Cancer Research UK Centre, University of Southampton, Southampton, United Kingdom; and
| | - Emanuele Zucca
- Institute of Oncology Research, Bellinzona, Switzerland
- Division of Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Medical Oncology, University of Bern, Bern, Switzerland
| |
Collapse
|
208
|
Presurgical Identification of Uterine Smooth Muscle Malignancies through the Characteristic FDG Uptake Pattern on PET Scans. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:7890241. [PMID: 30018513 PMCID: PMC6029472 DOI: 10.1155/2018/7890241] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022]
Abstract
The unidentified presence of uterine smooth muscle malignancies poses a tremendous risk in women planning surgery for presumed benign leiomyomas. We sought to investigate whether preoperative FDG PET may be useful to identify leiomyosarcomas (LMS) and smooth muscle tumors of uncertain malignant potential (STUMP). Methods. We investigated patients with rapidly growing uterine masses which were suspected of being malignant on ultrasound or MRI. Among the 21 patients who underwent FDG PET, we identified 7 LMS, 1 STUMP, and 13 leiomyomas. PET-derived parameters and FDG uptake patterns were analyzed retrospectively. Results. The SUVmax values of LMS/STUMP (range: 3.7–11.8) were significantly higher than those observed in leiomyomas (range: 2.0–9.4; P=0.003) despite a significant overlap. The metabolic tumor/necrosis ratio was significantly higher in LMS/STUMP than in leiomyomas (P < 0.001), with no significant intergroup overlaps. All LMS/STUMP revealed a characteristic pattern of FDG uptake, identifying a specific “hollow ball” sign (corresponding to areas of coagulative tumor necrosis). In contrast, this sign was invariably absent in patients with leiomyomas. Conclusion. The characteristic FDG uptake pattern instead of SUV on PET images allows identifying LMS/STUMP in patients with rapidly growing uterine masses, avoiding the deleterious consequences of regular surgery for presumed benign leiomyomas.
Collapse
|
209
|
Intratumoral heterogeneity in 18F-FDG PET/CT by textural analysis in breast cancer as a predictive and prognostic subrogate. Ann Nucl Med 2018; 32:379-388. [DOI: 10.1007/s12149-018-1253-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/01/2018] [Indexed: 10/14/2022]
|
210
|
Traverso A, Wee L, Dekker A, Gillies R. Repeatability and Reproducibility of Radiomic Features: A Systematic Review. Int J Radiat Oncol Biol Phys 2018; 102:1143-1158. [PMID: 30170872 PMCID: PMC6690209 DOI: 10.1016/j.ijrobp.2018.05.053] [Citation(s) in RCA: 551] [Impact Index Per Article: 78.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 05/15/2018] [Accepted: 05/20/2018] [Indexed: 12/15/2022]
Abstract
Purpose: An ever-growing number of predictive models used to inform clinical decision making have included quantitative, computer-extracted imaging biomarkers, or “radiomic features.” Broadly generalizable validity of radiomics-assisted models may be impeded by concerns about reproducibility. We offer a qualitative synthesis of 41 studies that specifically investigated the repeatability and reproducibility of radiomic features, derived from a systematic review of published peer-reviewed literature. Methods and Materials: The PubMed electronic database was searched using combinations of the broad Haynes and Ingui filters along with a set of text words specific to cancer, radiomics (including texture analyses), reproducibility, and repeatability. This review has been reported in compliance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. From each full-text article, information was extracted regarding cancer type, class of radiomic feature examined, reporting quality of key processing steps, and statistical metric used to segregate stable features. Results: Among 624 unique records, 41 full-text articles were subjected to review. The studies primarily addressed non-small cell lung cancer and oropharyngeal cancer. Only 7 studies addressed in detail every methodologic aspect related to image acquisition, preprocessing, and feature extraction. The repeatability and reproducibility of radiomic features are sensitive at various degrees to processing details such as image acquisition settings, image reconstruction algorithm, digital image preprocessing, and software used to extract radiomic features. First-order features were overall more reproducible than shape metrics and textural features. Entropy was consistently reported as one of the most stable first-order features. There was no emergent consensus regarding either shape metrics or textural features; however, coarseness and contrast appeared among the least reproducible. Conclusions: Investigations of feature repeatability and reproducibility are currently limited to a small number of cancer types. Reporting quality could be improved regarding details of feature extraction software, digital image manipulation (preprocessing), and the cutoff value used to distinguish stable features. We offer a qualitative synthesis of 41 studies that specifically investigated the repeatability and reproducibility of radiomic features. The repeatability and reproducibility of radiomic features are sensitive at various degrees to image quality and to software used to extract radiomic features. Investigations of feature repeatability and reproducibility are currently limited to a small number of cancer types. No consensus was found regarding the most repeatable and reproducible features with respect to different settings.
Collapse
Affiliation(s)
- Alberto Traverso
- Department of Radiation Oncology, MAASTRO Clinic, Maastricht, The Netherlands; School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands.
| | - Leonard Wee
- Department of Radiation Oncology, MAASTRO Clinic, Maastricht, The Netherlands; School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology, MAASTRO Clinic, Maastricht, The Netherlands; School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | | |
Collapse
|
211
|
Presotto L, Bettinardi V, De Bernardi E, Belli M, Cattaneo G, Broggi S, Fiorino C. PET textural features stability and pattern discrimination power for radiomics analysis: An “ad-hoc” phantoms study. Phys Med 2018; 50:66-74. [DOI: 10.1016/j.ejmp.2018.05.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/10/2018] [Accepted: 05/25/2018] [Indexed: 10/16/2022] Open
|
212
|
Stein D, Goldberg N, Domachevsky L, Bernstine H, Nidam M, Abadi-Korek I, Guindy M, Sosna J, Groshar D. Quantitative biomarkers for liver metastases: comparison of MRI diffusion-weighted imaging heterogeneity index and fluorine-18-fluoro-deoxyglucose standardised uptake value in hybrid PET/MR. Clin Radiol 2018; 73:832.e17-832.e22. [PMID: 29859634 DOI: 10.1016/j.crad.2018.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 04/18/2018] [Indexed: 01/24/2023]
Abstract
AIM To investigate the ability of apparent diffusion coefficient (ADC) heterogeneity index to discriminate liver metastases (LM) from normal-appearing liver (NAL) tissue as compared to common magnetic resonance imaging (MRI) metrics, and to investigate its correlation with 2-[18F]-fluoro-2-deoxy-d-glucose (18F-FDG) positron-emission tomography (PET) standardised uptake value (SUV). MATERIALS AND METHODS Thirty-nine liver metastases in 24 oncology patients (13 women, 11 men; mean age 56±13 years) with proven LM from heterogeneous sources were evaluated on a PET/MRI system. Abdominal sequences included Dixon and diffusion-weighted imaging (DWI) protocols with simultaneous PET. Tissue heterogeneity was calculated using the coefficient of variance (CV) of the ADC, and compared in LM and in NAL tissue of the same volume in an adjacent portion of the liver. The correlations between various ADC measures and PET SUV in distinguishing LM from NAL were evaluated. RESULTS A good correlation was found between ADCcv and SUVpeak (r=0.712). Moderate inverse correlation was found between ADCmin and SUVpeak (r=-0.536), and a weak inverse correlation between ADCmean and SUVpeak (r=-0.273). There was a significant difference between LM and NAL when ADCcv (p<0.0001) and ADCmin (p=0.001) were used. Receiver operating characteristic (ROC) analysis of SUV, ADCcv, ADCmin, and ADCmean produced an AUC of 0.989, 0.900, 0.742, and 0.623 respectively. CONCLUSIONS The ADCcv index is a potential biomarker of LM with better correlation to 18F-FDG PET SUVpeak than conventional MRI metrics, and may serve to quantitatively discriminate between LM and NAL.
Collapse
Affiliation(s)
- D Stein
- Department of Nuclear Medicine, Assuta Medical Centers, Tel-Aviv, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - N Goldberg
- Department of Nuclear Medicine, Assuta Medical Centers, Tel-Aviv, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - L Domachevsky
- Department of Nuclear Medicine, Assuta Medical Centers, Tel-Aviv, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - H Bernstine
- Department of Nuclear Medicine, Assuta Medical Centers, Tel-Aviv, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - M Nidam
- Department of Nuclear Medicine, Assuta Medical Centers, Tel-Aviv, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - I Abadi-Korek
- Department of Nuclear Medicine, Assuta Medical Centers, Tel-Aviv, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - M Guindy
- Department of Nuclear Medicine, Assuta Medical Centers, Tel-Aviv, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - J Sosna
- Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - D Groshar
- Department of Nuclear Medicine, Assuta Medical Centers, Tel-Aviv, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
213
|
Wolsztynski E, O'Sullivan F, Keyes E, O'Sullivan J, Eary JF. Positron emission tomography-based assessment of metabolic gradient and other prognostic features in sarcoma. J Med Imaging (Bellingham) 2018; 5:024502. [PMID: 29845091 PMCID: PMC5967597 DOI: 10.1117/1.jmi.5.2.024502] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/30/2018] [Indexed: 11/14/2022] Open
Abstract
Intratumoral heterogeneity biomarkers derived from positron emission tomography (PET) imaging with fluorodeoxyglucose (FDG) are of interest for a number of cancers, including sarcoma. A range of radiomic texture variables, adapted from general methodologies for image analysis, has shown promise in the setting. In the context of sarcoma, our group introduced an alternative model-based approach to the measurement of heterogeneity. In this approach, the heterogeneity of a tumor is characterized by the extent to which the 3-D FDG uptake pattern deviates from a simple elliptically contoured structure. By using a nonparametric analysis of the uptake profile obtained from this spatial model, a variable assessing the metabolic gradient of the tumor is developed. The work explores the prognostic potential of this new variable in the context of FDG-PET imaging of sarcoma. A mature clinical series involving 197 patients, 88 of whom have complete time-to-death information, is used. Texture variables based on the imaging data are also evaluated in this series and a range of appropriate machine learning methodologies are then used to explore the complementary prognostic roles for structure and texture variables. We conclude that both texture-based and model-based variables can be combined to achieve enhanced prognostic assessments of outcome for patients with sarcoma based on FDG-PET imaging information.
Collapse
Affiliation(s)
| | | | - Eimear Keyes
- University College Cork, Statistics Department, Cork, Ireland
| | | | - Janet F Eary
- National Cancer Institute, Bethesda, Maryland, United States
| |
Collapse
|
214
|
Katsila T, Matsoukas MT, Patrinos GP, Kardamakis D. Pharmacometabolomics Informs Quantitative Radiomics for Glioblastoma Diagnostic Innovation. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 21:429-439. [PMID: 28816643 DOI: 10.1089/omi.2017.0087] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Applications of omics systems biology technologies have enormous promise for radiology and diagnostics in surgical fields. In this context, the emerging fields of radiomics (a systems scale approach to radiology using a host of technologies, including omics) and pharmacometabolomics (use of metabolomics for patient and disease stratification and guiding precision medicine) offer much synergy for diagnostic innovation in surgery, particularly in neurosurgery. This synthesis of omics fields and applications is timely because diagnostic accuracy in central nervous system tumors still challenges decision-making. Considering the vast heterogeneity in brain tumors, disease phenotypes, and interindividual variability in surgical and chemotherapy outcomes, we believe that diagnostic accuracy can be markedly improved by quantitative radiomics coupled to pharmacometabolomics and related health information technologies while optimizing economic costs of traditional diagnostics. In this expert review, we present an innovation analysis on a systems-level multi-omics approach toward diagnostic accuracy in central nervous system tumors. For this, we suggest that glioblastomas serve as a useful application paradigm. We performed a literature search on PubMed for articles published in English between 2006 and 2016. We used the search terms "radiomics," "glioblastoma," "biomarkers," "pharmacogenomics," "pharmacometabolomics," "pharmacometabonomics/pharmacometabolomics," "collaborative informatics," and "precision medicine." A list of the top 4 insights we derived from this literature analysis is presented in this study. For example, we found that (i) tumor grading needs to be better refined, (ii) diagnostic precision should be improved, (iii) standardization in radiomics is lacking, and (iv) quantitative radiomics needs to prove clinical implementation. We conclude with an interdisciplinary call to the metabolomics, pharmacy/pharmacology, radiology, and surgery communities that pharmacometabolomics coupled to information technologies (chemoinformatics tools, databases, collaborative systems) can inform quantitative radiomics, thus translating Big Data and information growth to knowledge growth, rational drug development and diagnostics innovation for glioblastomas, and possibly in other brain tumors.
Collapse
Affiliation(s)
- Theodora Katsila
- 1 Department of Pharmacy, School of Health Sciences, University of Patras , Patras, Greece
| | | | - George P Patrinos
- 1 Department of Pharmacy, School of Health Sciences, University of Patras , Patras, Greece .,2 Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University , Al Ain, United Arab Emirates
| | - Dimitrios Kardamakis
- 3 Department of Radiation Oncology, University of Patras Medical School , Patras, Greece
| |
Collapse
|
215
|
Berenguer R, Pastor-Juan MDR, Canales-Vázquez J, Castro-García M, Villas MV, Mansilla Legorburo F, Sabater S. Radiomics of CT Features May Be Nonreproducible and Redundant: Influence of CT Acquisition Parameters. Radiology 2018; 288:407-415. [PMID: 29688159 DOI: 10.1148/radiol.2018172361] [Citation(s) in RCA: 422] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Purpose To identify the reproducible and nonredundant radiomics features (RFs) for computed tomography (CT). Materials and Methods Two phantoms were used to test RF reproducibility by using test-retest analysis, by changing the CT acquisition parameters (hereafter, intra-CT analysis), and by comparing five different scanners with the same CT parameters (hereafter, inter-CT analysis). Reproducible RFs were selected by using the concordance correlation coefficient (as a measure of the agreement between variables) and the coefficient of variation (defined as the ratio of the standard deviation to the mean). Redundant features were grouped by using hierarchical cluster analysis. Results A total of 177 RFs including intensity, shape, and texture features were evaluated. The test-retest analysis showed that 91% (161 of 177) of the RFs were reproducible according to concordance correlation coefficient. Reproducibility of intra-CT RFs, based on coefficient of variation, ranged from 89.3% (151 of 177) to 43.1% (76 of 177) where the pitch factor and the reconstruction kernel were modified, respectively. Reproducibility of inter-CT RFs, based on coefficient of variation, also showed large material differences, from 85.3% (151 of 177; wood) to only 15.8% (28 of 177; polyurethane). Ten clusters were identified after the hierarchical cluster analysis and one RF per cluster was chosen as representative. Conclusion Many RFs were redundant and nonreproducible. If all the CT parameters are fixed except field of view, tube voltage, and milliamperage, then the information provided by the analyzed RFs can be summarized in only 10 RFs (each representing a cluster) because of redundancy.
Collapse
Affiliation(s)
- Roberto Berenguer
- From the Departments of Medical Physics (R.B.), Radiation Oncology (S.S., M.V.V.), and Radiology (M.d.R.P.J.), Complejo Hospitalario Universitario de Albacete (CHUA), C/ Hnos Falcó 37, 02006 Albacete, Spain; Renewable Energy Research Institute, University of Castilla-La Mancha, Albacete, Spain (J.C.V., M.C.G.); and Mansilla Diagnóstico por Imagen, Albacete, Spain (F.M.L.)
| | - María Del Rosario Pastor-Juan
- From the Departments of Medical Physics (R.B.), Radiation Oncology (S.S., M.V.V.), and Radiology (M.d.R.P.J.), Complejo Hospitalario Universitario de Albacete (CHUA), C/ Hnos Falcó 37, 02006 Albacete, Spain; Renewable Energy Research Institute, University of Castilla-La Mancha, Albacete, Spain (J.C.V., M.C.G.); and Mansilla Diagnóstico por Imagen, Albacete, Spain (F.M.L.)
| | - Jesús Canales-Vázquez
- From the Departments of Medical Physics (R.B.), Radiation Oncology (S.S., M.V.V.), and Radiology (M.d.R.P.J.), Complejo Hospitalario Universitario de Albacete (CHUA), C/ Hnos Falcó 37, 02006 Albacete, Spain; Renewable Energy Research Institute, University of Castilla-La Mancha, Albacete, Spain (J.C.V., M.C.G.); and Mansilla Diagnóstico por Imagen, Albacete, Spain (F.M.L.)
| | - Miguel Castro-García
- From the Departments of Medical Physics (R.B.), Radiation Oncology (S.S., M.V.V.), and Radiology (M.d.R.P.J.), Complejo Hospitalario Universitario de Albacete (CHUA), C/ Hnos Falcó 37, 02006 Albacete, Spain; Renewable Energy Research Institute, University of Castilla-La Mancha, Albacete, Spain (J.C.V., M.C.G.); and Mansilla Diagnóstico por Imagen, Albacete, Spain (F.M.L.)
| | - María Victoria Villas
- From the Departments of Medical Physics (R.B.), Radiation Oncology (S.S., M.V.V.), and Radiology (M.d.R.P.J.), Complejo Hospitalario Universitario de Albacete (CHUA), C/ Hnos Falcó 37, 02006 Albacete, Spain; Renewable Energy Research Institute, University of Castilla-La Mancha, Albacete, Spain (J.C.V., M.C.G.); and Mansilla Diagnóstico por Imagen, Albacete, Spain (F.M.L.)
| | - Francisco Mansilla Legorburo
- From the Departments of Medical Physics (R.B.), Radiation Oncology (S.S., M.V.V.), and Radiology (M.d.R.P.J.), Complejo Hospitalario Universitario de Albacete (CHUA), C/ Hnos Falcó 37, 02006 Albacete, Spain; Renewable Energy Research Institute, University of Castilla-La Mancha, Albacete, Spain (J.C.V., M.C.G.); and Mansilla Diagnóstico por Imagen, Albacete, Spain (F.M.L.)
| | - Sebastià Sabater
- From the Departments of Medical Physics (R.B.), Radiation Oncology (S.S., M.V.V.), and Radiology (M.d.R.P.J.), Complejo Hospitalario Universitario de Albacete (CHUA), C/ Hnos Falcó 37, 02006 Albacete, Spain; Renewable Energy Research Institute, University of Castilla-La Mancha, Albacete, Spain (J.C.V., M.C.G.); and Mansilla Diagnóstico por Imagen, Albacete, Spain (F.M.L.)
| |
Collapse
|
216
|
Tsujikawa T, Tsuyoshi H, Kanno M, Yamada S, Kobayashi M, Narita N, Kimura H, Fujieda S, Yoshida Y, Okazawa H. Selected PET radiomic features remain the same. Oncotarget 2018; 9:20734-20746. [PMID: 29755685 PMCID: PMC5945508 DOI: 10.18632/oncotarget.25070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/24/2018] [Indexed: 01/12/2023] Open
Abstract
Purpose We investigated whether PET radiomic features are affected by differences in the scanner, scan protocol, and lesion location using 18F-FDG PET/CT and PET/MR scans. Results SUV, TMR, skewness, kurtosis, entropy, and homogeneity strongly correlated between PET/CT and PET/MR images. SUVs were significantly higher on PET/MR0-2 min and PET/MR0-10 min than on PET/CT in gynecological cancer (p = 0.008 and 0.008, respectively), whereas no significant difference was observed between PET/CT, PET/MR0–2 min, and PET/MR0–10 min images in oral cavity/oropharyngeal cancer. TMRs on PET/CT, PET/MR0–2 min, and PET/MR0–10 min increased in this order in gynecological cancer and oral cavity/oropharyngeal cancer. In contrast to conventional and histogram indices, 4 textural features (entropy, homogeneity, SRE, and LRE) were not significantly different between PET/CT, PET/MR0–2 min, and PET/MR0–10 min images. Conclusions 18F-FDG PET radiomic features strongly correlated between PET/CT and PET/MR images. Dixon-based attenuation correction on PET/MR images underestimated tumor tracer uptake more significantly in oral cavity/oropharyngeal cancer than in gynecological cancer. 18F-FDG PET textural features were affected less by differences in the scanner and scan protocol than conventional and histogram features, possibly due to the resampling process using a medium bin width. Methods Eight patients with gynecological cancer and 7 with oral cavity/oropharyngeal cancer underwent a whole-body 18F-FDG PET/CT scan and regional PET/MR scan in one day. PET/MR scans were performed for 10 minutes in the list mode, and PET/CT and 0–2 min and 0–10 min PET/MR images were reconstructed. The standardized uptake value (SUV), tumor-to-muscle SUV ratio (TMR), skewness, kurtosis, entropy, homogeneity, short-run emphasis (SRE), and long-run emphasis (LRE) were compared between PET/CT, PET/MR0-2 min, and PET/MR0-10 min images.
Collapse
Affiliation(s)
- Tetsuya Tsujikawa
- Biomedical Imaging Research Center, University of Fukui, Fukui, Japan
| | - Hideaki Tsuyoshi
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Masafumi Kanno
- Department of Otolaryngology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Shizuka Yamada
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Masato Kobayashi
- Wellness Promotion Science Center, College of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Norihiko Narita
- Department of Otolaryngology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hirohiko Kimura
- Department of Radiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Shigeharu Fujieda
- Department of Otolaryngology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yoshio Yoshida
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hidehiko Okazawa
- Biomedical Imaging Research Center, University of Fukui, Fukui, Japan
| |
Collapse
|
217
|
Wang S, Meng M, Zhang X, Wu C, Wang R, Wu J, Sami MU, Xu K. Texture analysis of diffusion weighted imaging for the evaluation of glioma heterogeneity based on different regions of interest. Oncol Lett 2018; 15:7297-7304. [PMID: 29731887 DOI: 10.3892/ol.2018.8232] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 02/23/2018] [Indexed: 12/21/2022] Open
Abstract
The present study aimed to explore the role of texture analysis with apparent diffusion coefficient (ADC) maps based on different regions of interest (ROI) in determining glioma grade. Thirty patients with glioma underwent diffusion-weighted imaging (DWI). ADC values were determined from the following three ROIs: i) whole tumor; ii) solid portion; and iii) peritumoral edema. Texture features were compared between high-grade gliomas (HGGs) and low-grade gliomas (LGGs) using the non-parametric Wilcoxon rank-sum test or the unpaired Student's t-test. Receiver operating characteristic (ROC) curves were constructed to determine the optimum threshold for inhomogeneity values in discrimination of HGGs from LGGs. With a spearman rank correlation model, the aforementioned ADC inhomogeneity values were correlated with the Ki-67 labeling index. With whole tumor ROI, inhomogeneity values proved to be significantly different between HGGs and LGGs (P<0.001). With solid portion ROI, inhomogeneity and median values showed significant difference between HGGs and LGGs (P=0.001 and P=0.043, respectively). With peritumoral edema ROI, entropy and edema volume demonstrated positive results (P=0.016, P<0.001). The whole tumor inhomogeneity parameter performed with better diagnostic accuracy (P=0.048) than selecting the solid portion ROI. The association between inhomogeneity and Ki-67 labeling index was significantly positive in whole tumor and solid portion ROI (R=0.628, P<0.001 and R=0.470, P=0.009). Texture analysis of DWI based on different ROI can provide various significant parameters to evaluate tumor heterogeneity, which were correlated with tumor grade. Particularly, the inhomogeneity value derived from whole tumor ROI provided high diagnostic value and predicting the status of tumor proliferation.
Collapse
Affiliation(s)
- Shan Wang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China.,Department of Radiology, Jiangsu Jiangyin People's Hospital, Jiangyin, Jiangsu 214400, P.R. China
| | - Meng Meng
- School of Medical Imaging, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Xue Zhang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Chen Wu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Ru Wang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Jiangfen Wu
- GE Healthcare (Shanghai) Co., Ltd., Shanghai 201203, P.R. China
| | - Muhammad Umair Sami
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Kai Xu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China.,Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, P.R. China
| |
Collapse
|
218
|
Bae JM, Jeong JY, Lee HY, Sohn I, Kim HS, Son JY, Kwon OJ, Choi JY, Lee KS, Shim YM. Pathologic stratification of operable lung adenocarcinoma using radiomics features extracted from dual energy CT images. Oncotarget 2018; 8:523-535. [PMID: 27880938 PMCID: PMC5352175 DOI: 10.18632/oncotarget.13476] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/14/2016] [Indexed: 02/07/2023] Open
Abstract
PURPOSE To evaluate the usefulness of surrogate biomarkers as predictors of histopathologic tumor grade and aggressiveness using radiomics data from dual-energy computed tomography (DECT), with the ultimate goal of accomplishing stratification of early-stage lung adenocarcinoma for optimal treatment. RESULTS Pathologic grade was divided into grades 1, 2, and 3. Multinomial logistic regression analysis revealed i-uniformity and 97.5th percentile CT attenuation value as independent significant factors to stratify grade 2 or 3 from grade 1. The AUC value calculated from leave-one-out cross-validation procedure for discriminating grades 1, 2, and 3 was 0.9307 (95% CI: 0.8514-1), 0.8610 (95% CI: 0.7547-0.9672), and 0.8394 (95% CI: 0.7045-0.9743), respectively. MATERIALS AND METHODS A total of 80 patients with 91 clinically and radiologically suspected stage I or II lung adenocarcinoma were prospectively enrolled. All patients underwent DECT and F-18-fluorodeoxyglucose (FDG) positron emission tomography (PET)/CT, followed by surgery. Quantitative CT and PET imaging characteristics were evaluated using a radiomics approach. Significant features for a tumor aggressiveness prediction model were extracted and used to calculate diagnostic performance for predicting all pathologic grades. CONCLUSIONS Quantitative radiomics values from DECT imaging metrics can help predict pathologic aggressiveness of lung adenocarcinoma.
Collapse
Affiliation(s)
- Jung Min Bae
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Ji Yun Jeong
- Department of Pathology, Kyungpook National University Medical Center, Kyungpook National University School of Medicine, Daegu 702-210, Korea
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Insuk Sohn
- Biostatistics and Clinical Epidemiology Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Hye Seung Kim
- Biostatistics and Clinical Epidemiology Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Ji Ye Son
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - O Jung Kwon
- Division of Respiratory and Critical Medicine of the Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Kyung Soo Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Young Mog Shim
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul 135-710, Korea
| |
Collapse
|
219
|
Niu XK, Chen ZF, Chen L, Li J, Peng T, Li X. Clinical Application of Biparametric MRI Texture Analysis for Detection and Evaluation of High-Grade Prostate Cancer in Zone-Specific Regions. AJR Am J Roentgenol 2018; 210:549-556. [PMID: 29220213 DOI: 10.2214/ajr.17.18494] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE The purpose of this study was to investigate the performance of biparametric MRI texture analysis (TA) in detecting and evaluating high-grade prostate cancer in zone-specific regions. MATERIALS AND METHODS A retrospective study included 184 consecutively registered biopsy-naive patients in whom prostate cancer was suspected who were undergoing multiparametric prostate MRI. MR images were scored and evaluated by two readers using the Prostate Imaging Reporting and Data System version 2 (PI-RADSv2) and biparametric MRI TA in separate sessions. Interobserver agreement on PI-RADSv2 score and textural parameters of biparametric MRI was evaluated. The logistic regression model based on TA was built for different zones of the prostate. ROC analysis was used to compare the TA-based model with other parameters alone. The correlation of each parameter with Gleason score of high-grade prostate cancer was also assessed. RESULTS Reader reliability ranged from moderate to good for PI-RADSv2 (Cohen κ = 0.525-0.616) and from good to excellent for textural metrics (intraclass correlation coefficient, 0.745-0.925). Diagnostic performance was significantly improved by use of the TA-based model (transition zone AUC, 0.87; peripheral zone AUC, 0.89) in comparison with PI-RADSv2 and other texture parameters alone. For the transition zone, entropy had moderate to good correlation with the Gleason score of high-grade prostate cancer (r = 0.562, p = 0.004). In the peripheral zone, entropy (r = 0.614, p = 0.003) and inertia (r = 0.663, p = 0.002) had moderate to good correlations with Gleason score. CONCLUSION The results of this clinical study indicate that a TA-based model that includes biparametric MRI can be used for identifying high-grade prostate cancer and that specific parameters extracted from TA may be additional tools for assessing tumor aggressiveness.
Collapse
Affiliation(s)
- Xiang-Ke Niu
- 1 Department of Radiology, Affiliated Hospital of Chengdu University, 82 2nd N Section of Second Ring Rd, Chengdu 610081, Sichuan, China
| | - Zhi-Fan Chen
- 1 Department of Radiology, Affiliated Hospital of Chengdu University, 82 2nd N Section of Second Ring Rd, Chengdu 610081, Sichuan, China
| | - Lin Chen
- 2 Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, China
| | - Jun Li
- 3 Department of General Surgery, Affiliated Hospital of Chengdu University, Chengdu, China
| | - Tao Peng
- 1 Department of Radiology, Affiliated Hospital of Chengdu University, 82 2nd N Section of Second Ring Rd, Chengdu 610081, Sichuan, China
| | - Xin Li
- 4 Life Science, Advanced Application Team, GE Healthcare, Beijing, China
| |
Collapse
|
220
|
Kebir S, Khurshid Z, Gaertner FC, Essler M, Hattingen E, Fimmers R, Scheffler B, Herrlinger U, Bundschuh RA, Glas M. Unsupervised consensus cluster analysis of [18F]-fluoroethyl-L-tyrosine positron emission tomography identified textural features for the diagnosis of pseudoprogression in high-grade glioma. Oncotarget 2018; 8:8294-8304. [PMID: 28030820 PMCID: PMC5352401 DOI: 10.18632/oncotarget.14166] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/23/2016] [Indexed: 02/07/2023] Open
Abstract
Rationale Timely detection of pseudoprogression (PSP) is crucial for the management of patients with high-grade glioma (HGG) but remains difficult. Textural features of O-(2-[18F]fluoroethyl)-L-tyrosine positron emission tomography (FET-PET) mirror tumor uptake heterogeneity; some of them may be associated with tumor progression. Methods Fourteen patients with HGG and suspected of PSP underwent FET-PET imaging. A set of 19 conventional and textural FET-PET features were evaluated and subjected to unsupervised consensus clustering. The final diagnosis of true progression vs. PSP was based on follow-up MRI using RANO criteria. Results Three robust clusters have been identified based on 10 predominantly textural FET-PET features. None of the patients with PSP fell into cluster 2, which was associated with high values for textural FET-PET markers of uptake heterogeneity. Three out of 4 patients with PSP were assigned to cluster 3 that was largely associated with low values of textural FET-PET features. By comparison, tumor-to-normal brain ratio (TNRmax) at the optimal cutoff 2.1 was less predictive of PSP (negative predictive value 57% for detecting true progression, p=0.07 vs. 75% with cluster 3, p=0.04). Principal Conclusions Clustering based on textural O-(2-[18F]fluoroethyl)-L-tyrosine PET features may provide valuable information in assessing the elusive phenomenon of pseudoprogression.
Collapse
Affiliation(s)
- Sied Kebir
- Division of Clinical Neurooncology, Department of Neurology, University of Bonn Medical Center, Germany.,Stem Cell Pathologies Group, Institute of Reconstructive Neurobiology, University of Bonn Medical Center, Germany
| | - Zain Khurshid
- Department of Nuclear Medicine, University of Bonn Medical Center, Germany
| | - Florian C Gaertner
- Department of Nuclear Medicine, University of Bonn Medical Center, Germany
| | - Markus Essler
- Department of Nuclear Medicine, University of Bonn Medical Center, Germany
| | - Elke Hattingen
- Neuroradiology, Department of Radiology, University of Bonn Medical Center, Germany
| | - Rolf Fimmers
- Institute of Medical Biometry, Informatics and Epidemiology, University of Bonn Medical Center, Germany
| | - Björn Scheffler
- Stem Cell Pathologies Group, Institute of Reconstructive Neurobiology, University of Bonn Medical Center, Germany.,DKFZ -Division of Translational Oncology/Neurooncology, German Cancer Consortium (DKTK) & University Hospital Essen, Germany
| | - Ulrich Herrlinger
- Division of Clinical Neurooncology, Department of Neurology, University of Bonn Medical Center, Germany
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, University of Bonn Medical Center, Germany
| | - Martin Glas
- Division of Clinical Neurooncology, Department of Neurology, University of Bonn Medical Center, Germany.,Stem Cell Pathologies Group, Institute of Reconstructive Neurobiology, University of Bonn Medical Center, Germany.,Clinical Cooperation Unit Neurooncology, MediClin Robert Janker Klinik, Bonn, Germany
| |
Collapse
|
221
|
Soufi M, Kamali-Asl A, Geramifar P, Rahmim A. A Novel Framework for Automated Segmentation and Labeling of Homogeneous Versus Heterogeneous Lung Tumors in [ 18F]FDG-PET Imaging. Mol Imaging Biol 2018; 19:456-468. [PMID: 27770402 DOI: 10.1007/s11307-016-1015-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE Determination of intra-tumor high-uptake area using 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET) imaging is an important consideration for dose painting in radiation treatment applications. The aim of our study was to develop a framework towards automated segmentation and labeling of homogeneous vs. heterogeneous tumors in clinical lung [18F]FDG-PET with the capability of intra-tumor high-uptake region delineation. PROCEDURES We utilized and extended a fuzzy random walk PET tumor segmentation algorithm to delineate intra-tumor high-uptake areas. Tumor textural feature (TF) analysis was used to find a relationship between tumor type and TF values. Segmentation accuracy was evaluated quantitatively utilizing 70 clinical [18F]FDG-PET lung images of patients with a total of 150 solid tumors. For volumetric analysis, the Dice similarity coefficient (DSC) and Hausdorff distance (HD) measures were extracted with respect to gold-standard manual segmentation. A multi-linear regression model was also proposed for automated tumor labeling based on TFs, including cross-validation analysis. RESULTS Two-tailed t test analysis of TFs between homogeneous and heterogeneous tumors revealed significant statistical difference for size-zone variability (SZV), intensity variability (IV), zone percentage (ZP), proposed parameters II and III, entropy and tumor volume (p < 0.001), dissimilarity, high intensity emphasis (HIE), and SUVmin (p < 0.01). Lower statistical differences were observed for proposed parameter I (p = 0.02), and no significant differences were observed for SUVmax and SUVmean. Furthermore, the Spearman rank analysis between visual tumor labeling and TF analysis depicted a significant correlation for SZV, IV, entropy, parameters II and III, and tumor volume (0.68 ≤ ρ ≤ 0.84) and moderate correlation for ZP, HIE, homogeneity, dissimilarity, parameter I, and SUVmin (0.22 ≤ ρ ≤ 0.52), while no correlations were observed for SUVmax and SUVmean (ρ < 0.08). The multi-linear regression model for automated tumor labeling process resulted in R 2 and RMSE values of 0.93 and 0.14, respectively (p < 0.001), and generated tumor labeling sensitivity and specificity of 0.93 and 0.89. With respect to baseline random walk segmentation, the results showed significant (p < 0.001) mean DSC, HD, and SUVmean error improvements of 21.4 ± 11.5 %, 1.4 ± 0.8 mm, and 16.8 ± 8.1 % in homogeneous tumors and 7.4 ± 4.4 %, 1.5 ± 0.6 mm, and 7.9 ± 2.7 % in heterogeneous lesions. In addition, significant (p < 0.001) mean DSC, HD, and SUVmean error improvements were observed for tumor sub-volume delineations, namely 5 ± 2 %, 1.5 ± 0.6 mm, and 7 ± 3 % for the proposed Fuzzy RW method compared to RW segmentation. CONCLUSION We proposed and demonstrated an automatic framework for significantly improved segmentation and labeling of homogeneous vs. heterogeneous tumors in lung [18F]FDG-PET images.
Collapse
Affiliation(s)
- Motahare Soufi
- Department of Radiation Medicine Engineering, Shahid Beheshti University, Tehran, Iran
| | - Alireza Kamali-Asl
- Department of Radiation Medicine Engineering, Shahid Beheshti University, Tehran, Iran.
| | - Parham Geramifar
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Arman Rahmim
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Electrical & Computer Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
222
|
Werner RA, Lapa C, Ilhan H, Higuchi T, Buck AK, Lehner S, Bartenstein P, Bengel F, Schatka I, Muegge DO, Papp L, Zsótér N, Große-Ophoff T, Essler M, Bundschuh RA. Survival prediction in patients undergoing radionuclide therapy based on intratumoral somatostatin-receptor heterogeneity. Oncotarget 2018; 8:7039-7049. [PMID: 27705948 PMCID: PMC5351689 DOI: 10.18632/oncotarget.12402] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 09/26/2016] [Indexed: 11/25/2022] Open
Abstract
The NETTER-1 trial demonstrated significantly improved progression-free survival (PFS) for peptide receptor radionuclide therapy (PRRT) in neuroendocrine tumors (NET) emphasizing the high demand for response prediction in appropriate candidates. In this multicenter study, we aimed to elucidate the prognostic value of tumor heterogeneity as assessed by somatostatin receptor (SSTR)-PET/CT. 141 patients with SSTR-expressing tumors were analyzed obtaining SSTR-PET/CT before PRRT (1-6 cycles, 177Lu somatostatin analog). Using the Interview Fusion Workstation (Mediso), a total of 872 metastases were manually segmented. Conventional PET parameters as well as textural features representing intratumoral heterogeneity were computed. The prognostic ability for PFS and overall survival (OS) were examined. After performing Cox regression, independent parameters were determined by ROC analysis to obtain cut-off values to be used for Kaplan-Meier analysis. Within follow-up (median, 43.1 months), 75 patients showed disease progression (median, 22.2 m) and 54 patients died (median, 27.6 m). Cox analysis identified 8 statistically independent heterogeneity parameters for time-to-progression and time-to-death. Among them, the textural feature Entropy predicted both PFS and OS. Conventional PET parameters failed in response prediction. Imaging-based heterogeneity assessment provides prognostic information in PRRT candidates and outperformed conventional PET parameters. Its implementation in clinical practice can pave the way for individualized patient management.
Collapse
Affiliation(s)
- Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Harun Ilhan
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Sebastian Lehner
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Frank Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - László Papp
- Department of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Tobias Große-Ophoff
- Department of Nuclear Medicine, University Medical Center Bonn, Bonn, Germany
| | - Markus Essler
- Department of Nuclear Medicine, University Medical Center Bonn, Bonn, Germany
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, University Medical Center Bonn, Bonn, Germany
| |
Collapse
|
223
|
Ben Bouallègue F, Vauchot F, Mariano-Goulart D, Payoux P. Diagnostic and prognostic value of amyloid PET textural and shape features: comparison with classical semi-quantitative rating in 760 patients from the ADNI-2 database. Brain Imaging Behav 2018; 13:111-125. [PMID: 29427064 DOI: 10.1007/s11682-018-9833-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We evaluated the performance of amyloid PET textural and shape features in discriminating normal and Alzheimer's disease (AD) subjects, and in predicting conversion to AD in subjects with mild cognitive impairment (MCI) or significant memory concern (SMC). Subjects from the Alzheimer's Disease Neuroimaging Initiative with available baseline 18F-florbetapir and T1-MRI scans were included. The cross-sectional cohort consisted of 181 controls and 148 AD subjects. The longitudinal cohort consisted of 431 SMC/MCI subjects, 85 of whom converted to AD during follow-up. PET images were normalized to MNI space and post-processed using in-house software. Relative retention indices (SUVr) were computed with respect to pontine, cerebellar, and composite reference regions. Several textural and shape features were extracted then combined using a support vector machine (SVM) to build a predictive model of AD conversion. Diagnostic and prognostic performance was evaluated using ROC analysis and survival analysis with the Cox proportional hazard model. The three SUVr and all the tested features effectively discriminated AD subjects in cross-sectional analysis (all p < 0.001). In longitudinal analysis, the variables with the highest prognostic value were composite SUVr (AUC 0.86; accuracy 81%), skewness (0.87; 83%), local minima (0.85; 79%), Geary's index (0.86; 81%), gradient norm maximal argument (0.83; 82%), and the SVM model (0.91; 86%). The adjusted hazard ratio for AD conversion was 5.5 for the SVM model, compared with 4.0, 2.6, and 3.8 for cerebellar, pontine and composite SUVr (all p < 0.001), indicating that appropriate amyloid textural and shape features predict conversion to AD with at least as good accuracy as classical SUVr.
Collapse
Affiliation(s)
- Fayçal Ben Bouallègue
- Nuclear Medicine Department, Montpellier University Hospital, Montpellier, France. .,Nuclear Medicine Department, Purpan University Hospital, Toulouse, France.
| | - Fabien Vauchot
- Nuclear Medicine Department, Montpellier University Hospital, Montpellier, France
| | - Denis Mariano-Goulart
- Nuclear Medicine Department, Montpellier University Hospital, Montpellier, France.,PhyMedExp, INSERM - CNRS, Montpellier University, Montpellier, France
| | - Pierre Payoux
- Nuclear Medicine Department, Purpan University Hospital, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| |
Collapse
|
224
|
Xia W, Chen Y, Zhang R, Yan Z, Zhou X, Zhang B, Gao X. Radiogenomics of hepatocellular carcinoma: multiregion analysis-based identification of prognostic imaging biomarkers by integrating gene data-a preliminary study. Phys Med Biol 2018; 63:035044. [PMID: 29311419 DOI: 10.1088/1361-6560/aaa609] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Our objective was to identify prognostic imaging biomarkers for hepatocellular carcinoma in contrast-enhanced computed tomography (CECT) with biological interpretations by associating imaging features and gene modules. We retrospectively analyzed 371 patients who had gene expression profiles. For the 38 patients with CECT imaging data, automatic intra-tumor partitioning was performed, resulting in three spatially distinct subregions. We extracted a total of 37 quantitative imaging features describing intensity, geometry, and texture from each subregion. Imaging features were selected after robustness and redundancy analysis. Gene modules acquired from clustering were chosen for their prognostic significance. By constructing an association map between imaging features and gene modules with Spearman rank correlations, the imaging features that significantly correlated with gene modules were obtained. These features were evaluated with Cox's proportional hazard models and Kaplan-Meier estimates to determine their prognostic capabilities for overall survival (OS). Eight imaging features were significantly correlated with prognostic gene modules, and two of them were associated with OS. Among these, the geometry feature volume fraction of the subregion, which was significantly correlated with all prognostic gene modules representing cancer-related interpretation, was predictive of OS (Cox p = 0.022, hazard ratio = 0.24). The texture feature cluster prominence in the subregion, which was correlated with the prognostic gene module representing lipid metabolism and complement activation, also had the ability to predict OS (Cox p = 0.021, hazard ratio = 0.17). Imaging features depicting the volume fraction and textural heterogeneity in subregions have the potential to be predictors of OS with interpretable biological meaning.
Collapse
Affiliation(s)
- Wei Xia
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, 88 Keling Rd, Suzhou 215163, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
225
|
Shaikh F, Franc B, Allen E, Sala E, Awan O, Hendrata K, Halabi S, Mohiuddin S, Malik S, Hadley D, Shrestha R. Translational Radiomics: Defining the Strategy Pipeline and Considerations for Application-Part 2: From Clinical Implementation to Enterprise. J Am Coll Radiol 2018; 15:543-549. [PMID: 29366598 DOI: 10.1016/j.jacr.2017.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 12/07/2017] [Indexed: 12/18/2022]
Abstract
Enterprise imaging has channeled various technological innovations to the field of clinical radiology, ranging from advanced imaging equipment and postacquisition iterative reconstruction tools to image analysis and computer-aided detection tools. More recently, the advancement in the field of quantitative image analysis coupled with machine learning-based data analytics, classification, and integration has ushered in the era of radiomics, a paradigm shift that holds tremendous potential in clinical decision support as well as drug discovery. However, there are important issues to consider to incorporate radiomics into a clinically applicable system and a commercially viable solution. In this two-part series, we offer insights into the development of the translational pipeline for radiomics from methodology to clinical implementation (Part 1) and from that point to enterprise development (Part 2). In Part 2 of this two-part series, we study the components of the strategy pipeline, from clinical implementation to building enterprise solutions.
Collapse
Affiliation(s)
- Faiq Shaikh
- Institute of Computational Health Sciences, UCSF, San Francisco, California.
| | - Benjamin Franc
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California
| | | | - Evis Sala
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Omer Awan
- Department of Radiology, Temple University, Philadelphia, Pennsylvania
| | | | - Safwan Halabi
- Department of Radiology, Stanford University, Palo Alto, California
| | - Sohaib Mohiuddin
- Department of Radiology, Division of Nuclear Medicine, University of Miami, Miami, Florida
| | - Sana Malik
- School of Social Welfare, Stony Brook University, New York, New York
| | - Dexter Hadley
- Institute of Computational Health Sciences, UCSF, San Francisco, California
| | | |
Collapse
|
226
|
Zhou H, Dong D, Chen B, Fang M, Cheng Y, Gan Y, Zhang R, Zhang L, Zang Y, Liu Z, Zheng H, Li W, Tian J. Diagnosis of Distant Metastasis of Lung Cancer: Based on Clinical and Radiomic Features. Transl Oncol 2018; 11:31-36. [PMID: 29156383 PMCID: PMC5697996 DOI: 10.1016/j.tranon.2017.10.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/24/2017] [Accepted: 10/26/2017] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES To analyze the distant metastasis possibility based on computed tomography (CT) radiomic features in patients with lung cancer. METHODS This was a retrospective analysis of 348 patients with lung cancer enrolled between 2014 and February 2015. A feature set containing clinical features and 485 radiomic features was extracted from the pretherapy CT images. Feature selection via concave minimization (FSV) was used to select effective features. A support vector machine (SVM) was used to evaluate the predictive ability of each feature. RESULTS Four radiomic features and three clinical features were obtained by FSV feature selection. Classification accuracy by the proposed SVM with SGD method was 71.02%, and the area under the curve was 72.84% with only the radiomic features extracted from CT. After the addition of clinical features, 89.09% can be achieved. CONCLUSION The radiomic features of the pretherapy CT images may be used as predictors of distant metastasis. And it also can be used in combination with the patient's gender and tumor T and N phase information to diagnose the possibility of distant metastasis in lung cancer.
Collapse
Affiliation(s)
- Hongyu Zhou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave., SZ University Town, Shenzhen, China, 518055; CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing, China
| | - Di Dong
- CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing, China
| | - Bojiang Chen
- Department of respiratory and critical care medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Mengjie Fang
- CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yue Cheng
- Department of respiratory and critical care medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuncun Gan
- Department of respiratory and critical care medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rui Zhang
- Department of respiratory and critical care medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Liwen Zhang
- CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yali Zang
- CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhenyu Liu
- CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave., SZ University Town, Shenzhen, China, 518055.
| | - Weimin Li
- Department of respiratory and critical care medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Jie Tian
- CAS Key Lab of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
227
|
Park S, Ha S, Lee SH, Paeng JC, Keam B, Kim TM, Kim DW, Heo DS. Intratumoral heterogeneity characterized by pretreatment PET in non-small cell lung cancer patients predicts progression-free survival on EGFR tyrosine kinase inhibitor. PLoS One 2018; 13:e0189766. [PMID: 29385152 PMCID: PMC5791940 DOI: 10.1371/journal.pone.0189766] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 12/01/2017] [Indexed: 01/08/2023] Open
Abstract
Intratumoral heterogeneity has been suggested to be an important resistance mechanism leading to treatment failure. We hypothesized that radiologic images could be an alternative method for identification of tumor heterogeneity. We tested heterogeneity textural parameters on pretreatment FDG-PET/CT in order to assess the predictive value of target therapy. Recurred or metastatic non-small cell lung cancer (NSCLC) subjects with an activating EGFR mutation treated with either gefitinib or erlotinib were reviewed. An exploratory data set (n = 161) and a validation data set (n = 21) were evaluated, and eight parameters were selected for survival analysis. The optimal cutoff value was determined by the recursive partitioning method, and the predictive value was calculated using Harrell’s C-index. Univariate analysis revealed that all eight parameters showed an increased hazard ratio (HR) for progression-free survival (PFS). The highest HR was 6.41 (P<0.01) with co-occurrence (Co) entropy. Increased risk remained present after adjusting for initial stage, performance status (PS), and metabolic volume (MV) (aHR: 4.86, P<0.01). Textural parameters were found to have an incremental predictive value of early EGFR tyrosine kinase inhibitor (TKI) failure compared to that of the base model of the stage and PS (C-index 0.596 vs. 0.662, P = 0.02, by Co entropy). Heterogeneity textural parameters acquired from pretreatment FDG-PET/CT are highly predictive factors for PFS of EGFR TKI in EGFR-mutated NSCLC patients. These parameters are easily applicable to the identification of a subpopulation at increased risk of early EGFR TKI failure. Correlation to genomic alteration should be determined in future studies.
Collapse
Affiliation(s)
- Sehhoon Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seunggyun Ha
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University Hospital, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- * E-mail: (SHL); (JCP)
| | - Jin Chul Paeng
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University Hospital, Seoul, Republic of Korea
- * E-mail: (SHL); (JCP)
| | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong-Wan Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dae Seog Heo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
228
|
Wu L, Wang C, Tan X, Cheng Z, Zhao K, Yan L, Liang Y, Liu Z, Liang C. Radiomics approach for preoperative identification of stages I -II and III -IV of esophageal cancer. Chin J Cancer Res 2018; 30:396-405. [PMID: 30210219 PMCID: PMC6129566 DOI: 10.21147/j.issn.1000-9604.2018.04.02] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Objective To predict preoperative staging using a radiomics approach based on computed tomography (CT) images of patients with esophageal squamous cell carcinoma (ESCC). Methods This retrospective study included 154 patients (primary cohort: n=114; validation cohort: n=40) with pathologically confirmed ESCC. All patients underwent a preoperative CT scan from the neck to abdomen. High throughput and quantitative radiomics features were extracted from the CT images for each patient. A radiomics signature was constructed using the least absolute shrinkage and selection operator (Lasso). Associations between radiomics signature, tumor volume and ESCC staging were explored. Diagnostic performance of radiomics approach and tumor volume for discriminating between stages I-II and III-IV was evaluated and compared using the receiver operating characteristics (ROC) curves and net reclassification improvement (NRI). Results A total of 9,790 radiomics features were extracted. Ten features were selected to build a radiomics signature after feature dimension reduction. The radiomics signature was significantly associated with ESCC staging (P<0.001), and yielded a better performance for discrimination of early and advanced stage ESCC compared to tumor volume in both the primary [area under the receiver operating characteristic curve (AUC): 0.795vs. 0.694, P=0.003; NRI=0.424)] and validation cohorts (AUC: 0.762 vs. 0.624, P=0.035; NRI=0.834). Conclusions The quantitative approach has the potential to identify stage I-II and III-IV ESCC before treatment.
Collapse
Affiliation(s)
- Lei Wu
- School of Medicine, South China University of Technology, Guangzhou 510006, China.,Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Cong Wang
- School of Automation Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Xianzheng Tan
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Zixuan Cheng
- School of Medicine, South China University of Technology, Guangzhou 510006, China.,Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Ke Zhao
- School of Medicine, South China University of Technology, Guangzhou 510006, China.,Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Lifen Yan
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yanli Liang
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Changhong Liang
- School of Medicine, South China University of Technology, Guangzhou 510006, China.,Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| |
Collapse
|
229
|
Acharya UR, Hagiwara Y, Sudarshan VK, Chan WY, Ng KH. Towards precision medicine: from quantitative imaging to radiomics. J Zhejiang Univ Sci B 2018; 19:6-24. [PMID: 29308604 PMCID: PMC5802973 DOI: 10.1631/jzus.b1700260] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/16/2017] [Indexed: 12/12/2022]
Abstract
Radiology (imaging) and imaging-guided interventions, which provide multi-parametric morphologic and functional information, are playing an increasingly significant role in precision medicine. Radiologists are trained to understand the imaging phenotypes, transcribe those observations (phenotypes) to correlate with underlying diseases and to characterize the images. However, in order to understand and characterize the molecular phenotype (to obtain genomic information) of solid heterogeneous tumours, the advanced sequencing of those tissues using biopsy is required. Thus, radiologists image the tissues from various views and angles in order to have the complete image phenotypes, thereby acquiring a huge amount of data. Deriving meaningful details from all these radiological data becomes challenging and raises the big data issues. Therefore, interest in the application of radiomics has been growing in recent years as it has the potential to provide significant interpretive and predictive information for decision support. Radiomics is a combination of conventional computer-aided diagnosis, deep learning methods, and human skills, and thus can be used for quantitative characterization of tumour phenotypes. This paper discusses the overview of radiomics workflow, the results of various radiomics-based studies conducted using various radiological images such as computed tomography (CT), magnetic resonance imaging (MRI), and positron-emission tomography (PET), the challenges we are facing, and the potential contribution of radiomics towards precision medicine.
Collapse
Affiliation(s)
- U. Rajendra Acharya
- Department of Electronics and Computer Engineering, Ngee Ann Polytechnic, Singapore 599489, Singapore
- Department of Biomedical Engineering, School of Science and Technology, Singapore University of Social Sciences, Singapore 599494, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Yuki Hagiwara
- Department of Electronics and Computer Engineering, Ngee Ann Polytechnic, Singapore 599489, Singapore
| | - Vidya K. Sudarshan
- Department of Electronics and Computer Engineering, Ngee Ann Polytechnic, Singapore 599489, Singapore
| | - Wai Yee Chan
- Department of Biomedical Imaging, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Kwan Hoong Ng
- Department of Biomedical Imaging, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
230
|
Investigating the Robustness Neighborhood Gray Tone Difference Matrix and Gray Level Co-occurrence Matrix Radiomic Features on Clinical Computed Tomography Systems Using Anthropomorphic Phantoms: Evidence From a Multivendor Study. J Comput Assist Tomogr 2017; 41:995-1001. [PMID: 28708732 DOI: 10.1097/rct.0000000000000632] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The aim of this study was to determine if optimized imaging protocols across multiple computed tomography (CT) vendors could result in reproducible radiomic features calculated from an anthropomorphic phantom. METHODS Materials with varying degrees of heterogeneity were placed throughout the lungs of the phantom. Twenty scans of the phantom were acquired on 3 CT manufacturers with chest CT protocols that had optimized protocol parameters. Scans were reconstructed using vendor-specific standards and lung kernels. The concordance correlation coefficient (CCC) was used to calculate reproducibility between features. For features with high CCC values, Bland-Altman analysis was also used to quantify agreement. RESULTS The mean Hounsfield unit (HU) was 32.93 HU (141.7 to -26.5 HU) for the rubber insert and 347.2 HU (-320.9 to -347.7 HU) for the wood insert. Low CCC values of less than 0.9 were calculated for all features across all scans. CONCLUSIONS Radiomic features that are derived from the spatial distribution of voxel intensities should be particularly scrutinized for reproducibility in a multivendor environment.
Collapse
|
231
|
Wang H, Zhou Z, Li Y, Chen Z, Lu P, Wang W, Liu W, Yu L. Comparison of machine learning methods for classifying mediastinal lymph node metastasis of non-small cell lung cancer from 18F-FDG PET/CT images. EJNMMI Res 2017; 7:11. [PMID: 28130689 PMCID: PMC5272853 DOI: 10.1186/s13550-017-0260-9] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/19/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND This study aimed to compare one state-of-the-art deep learning method and four classical machine learning methods for classifying mediastinal lymph node metastasis of non-small cell lung cancer (NSCLC) from 18F-FDG PET/CT images. Another objective was to compare the discriminative power of the recently popular PET/CT texture features with the widely used diagnostic features such as tumor size, CT value, SUV, image contrast, and intensity standard deviation. The four classical machine learning methods included random forests, support vector machines, adaptive boosting, and artificial neural network. The deep learning method was the convolutional neural networks (CNN). The five methods were evaluated using 1397 lymph nodes collected from PET/CT images of 168 patients, with corresponding pathology analysis results as gold standard. The comparison was conducted using 10 times 10-fold cross-validation based on the criterion of sensitivity, specificity, accuracy (ACC), and area under the ROC curve (AUC). For each classical method, different input features were compared to select the optimal feature set. Based on the optimal feature set, the classical methods were compared with CNN, as well as with human doctors from our institute. RESULTS For the classical methods, the diagnostic features resulted in 81~85% ACC and 0.87~0.92 AUC, which were significantly higher than the results of texture features. CNN's sensitivity, specificity, ACC, and AUC were 84, 88, 86, and 0.91, respectively. There was no significant difference between the results of CNN and the best classical method. The sensitivity, specificity, and ACC of human doctors were 73, 90, and 82, respectively. All the five machine learning methods had higher sensitivities but lower specificities than human doctors. CONCLUSIONS The present study shows that the performance of CNN is not significantly different from the best classical methods and human doctors for classifying mediastinal lymph node metastasis of NSCLC from PET/CT images. Because CNN does not need tumor segmentation or feature calculation, it is more convenient and more objective than the classical methods. However, CNN does not make use of the import diagnostic features, which have been proved more discriminative than the texture features for classifying small-sized lymph nodes. Therefore, incorporating the diagnostic features into CNN is a promising direction for future research.
Collapse
Affiliation(s)
- Hongkai Wang
- Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, No. 2 Linggong Street, Ganjingzi District, Dalian, Liaoning, 116024, China
| | - Zongwei Zhou
- Department of Biomedical Informatics and the College of Health Solutions, Arizona State University, 13212 East Shea Boulevard, Scottsdale, AZ, 85259, USA
| | - Yingci Li
- Center of PET/CT, The Affiliated Tumor Hospital of Harbin Medical University, 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Zhonghua Chen
- Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, No. 2 Linggong Street, Ganjingzi District, Dalian, Liaoning, 116024, China
| | - Peiou Lu
- Center of PET/CT, The Affiliated Tumor Hospital of Harbin Medical University, 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Wenzhi Wang
- Center of PET/CT, The Affiliated Tumor Hospital of Harbin Medical University, 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Wanyu Liu
- HIT-INSA Sino French Research Centre for Biomedical Imaging, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Lijuan Yu
- Center of PET/CT, The Affiliated Tumor Hospital of Harbin Medical University, 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, China.
| |
Collapse
|
232
|
Lovat E, Siddique M, Goh V, Ferner RE, Cook GJR, Warbey VS. The effect of post-injection 18F-FDG PET scanning time on texture analysis of peripheral nerve sheath tumours in neurofibromatosis-1. EJNMMI Res 2017; 7:35. [PMID: 28429332 PMCID: PMC5399011 DOI: 10.1186/s13550-017-0282-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 04/04/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Texture features are being increasingly evaluated in 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) as adjunctive imaging biomarkers in a number of different cancers. Whilst studies have reported repeatability between scans, there have been no studies that have specifically investigated the effect that the time of acquisition post-injection of 18F-FDG has on texture features. The aim of this study was to investigate if texture features change between scans performed at different time points post-injection. RESULTS Fifty-four patients (30 male, 24 female, mean age 35.1 years) with neurofibromatosis-1 and suspected malignant transformation of a neurofibroma underwent 18F-FDG PET/computed tomography (CT) scans at 101.5 ± 15.0 and 251.7 ± 18.4 min post-injection of 350 MBq 18F-FDG to a standard clinical protocol. Following tumour segmentation on both early and late scans, first- (n = 37), second- (n = 25) and high-order (n = 31) statistical features, as well as fractal texture features (n = 6), were calculated and a comparison was made between the early and late scans for each feature. Of the 54 tumours, 30 were benign and 24 malignant on histological analysis or on clinical follow-up for at least 5 years. Overall, 25/37 first-order, 9/25 second-order, 13/31 high-order and 3/6 fractal features changed significantly (p < 0.05) between early and late scans. The corresponding proportions for the 30 benign tumours alone were 22/37, 7/25, 8/31 and 2/6 and for the 24 malignant tumours, 11/37, 6/25, 8/31 and 0/6, respectively. CONCLUSIONS Several texture features change with time post-injection of 18F-FDG. Thus, when comparing texture features in intra- and inter-patient studies, it is essential that scans are obtained at a consistent time post-injection of 18F-FDG.
Collapse
Affiliation(s)
- Eitan Lovat
- Guy's, King's and St Thomas' School of Medicine, King's College London, London, UK
| | - Musib Siddique
- Department of Cancer Imaging, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| | - Vicky Goh
- Department of Cancer Imaging, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| | - Rosalie E Ferner
- National Neurofibromatosis Service, Department of Neurology, Guys and St Thomas' NHS Foundation Trust, London, UK
| | - Gary J R Cook
- Department of Cancer Imaging, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK.
- Clinical PET Centre, Division of Imaging Sciences and Biomedical Engineering, King's College London, St Thomas' Hospital, London, SE1 7EH, UK.
| | - Victoria S Warbey
- Department of Cancer Imaging, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| |
Collapse
|
233
|
Bashir U, Azad G, Siddique MM, Dhillon S, Patel N, Bassett P, Landau D, Goh V, Cook G. The effects of segmentation algorithms on the measurement of 18F-FDG PET texture parameters in non-small cell lung cancer. EJNMMI Res 2017; 7:60. [PMID: 28748524 PMCID: PMC5529305 DOI: 10.1186/s13550-017-0310-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/20/2017] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Measures of tumour heterogeneity derived from 18-fluoro-2-deoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) scans are increasingly reported as potential biomarkers of non-small cell lung cancer (NSCLC) for classification and prognostication. Several segmentation algorithms have been used to delineate tumours, but their effects on the reproducibility and predictive and prognostic capability of derived parameters have not been evaluated. The purpose of our study was to retrospectively compare various segmentation algorithms in terms of inter-observer reproducibility and prognostic capability of texture parameters derived from non-small cell lung cancer (NSCLC) 18F-FDG PET/CT images. Fifty three NSCLC patients (mean age 65.8 years; 31 males) underwent pre-chemoradiotherapy 18F-FDG PET/CT scans. Three readers segmented tumours using freehand (FH), 40% of maximum intensity threshold (40P), and fuzzy locally adaptive Bayesian (FLAB) algorithms. Intraclass correlation coefficient (ICC) was used to measure the inter-observer variability of the texture features derived by the three segmentation algorithms. Univariate cox regression was used on 12 commonly reported texture features to predict overall survival (OS) for each segmentation algorithm. Model quality was compared across segmentation algorithms using Akaike information criterion (AIC). RESULTS 40P was the most reproducible algorithm (median ICC 0.9; interquartile range [IQR] 0.85-0.92) compared with FLAB (median ICC 0.83; IQR 0.77-0.86) and FH (median ICC 0.77; IQR 0.7-0.85). On univariate cox regression analysis, 40P found 2 out of 12 variables, i.e. first-order entropy and grey-level co-occurence matrix (GLCM) entropy, to be significantly associated with OS; FH and FLAB found 1, i.e., first-order entropy. For each tested variable, survival models for all three segmentation algorithms were of similar quality, exhibiting comparable AIC values with overlapping 95% CIs. CONCLUSIONS Compared with both FLAB and FH, segmentation with 40P yields superior inter-observer reproducibility of texture features. Survival models generated by all three segmentation algorithms are of at least equivalent utility. Our findings suggest that a segmentation algorithm using a 40% of maximum threshold is acceptable for texture analysis of 18F-FDG PET in NSCLC.
Collapse
Affiliation(s)
- Usman Bashir
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
| | - Gurdip Azad
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
| | - Muhammad Musib Siddique
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
| | - Saana Dhillon
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
| | - Nikheel Patel
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
| | - Paul Bassett
- Stats Consultancy Ltd, 40 Longwood Lane, Amersham, Bucks HP7 9EN UK
| | - David Landau
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
- Department of Clinical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London, SE1 9RT UK
| | - Vicky Goh
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
- Department of Radiology, Guy’s Hospital, 2nd Floor, Tower Wing, Great Maze Pond, London, SE1 9RT UK
| | - Gary Cook
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
- PET Imaging Centre and the Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
| |
Collapse
|
234
|
Lapa P, Marques M, Isidoro J, Barata F, Costa G, de Lima J. 18 F-FDG PET/CT in lung cancer. The added value of quantification. Rev Esp Med Nucl Imagen Mol 2017. [DOI: 10.1016/j.remnie.2017.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
235
|
Phillips I, Ajaz M, Ezhil V, Prakash V, Alobaidli S, McQuaid SJ, South C, Scuffham J, Nisbet A, Evans P. Clinical applications of textural analysis in non-small cell lung cancer. Br J Radiol 2017; 91:20170267. [PMID: 28869399 DOI: 10.1259/bjr.20170267] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the leading cause of cancer mortality worldwide. Treatment pathways include regular cross-sectional imaging, generating large data sets which present intriguing possibilities for exploitation beyond standard visual interpretation. This additional data mining has been termed "radiomics" and includes semantic and agnostic approaches. Textural analysis (TA) is an example of the latter, and uses a range of mathematically derived features to describe an image or region of an image. Often TA is used to describe a suspected or known tumour. TA is an attractive tool as large existing image sets can be submitted to diverse techniques for data processing, presentation, interpretation and hypothesis testing with annotated clinical outcomes. There is a growing anthology of published data using different TA techniques to differentiate between benign and malignant lung nodules, differentiate tissue subtypes of lung cancer, prognosticate and predict outcome and treatment response, as well as predict treatment side effects and potentially aid radiotherapy planning. The aim of this systematic review is to summarize the current published data and understand the potential future role of TA in managing lung cancer.
Collapse
Affiliation(s)
- Iain Phillips
- 1 Royal Surrey County Hospital, University of Surrey, Guildford, UK
| | - Mazhar Ajaz
- 1 Royal Surrey County Hospital, University of Surrey, Guildford, UK.,2 Surrey Clinical Research Centre, University of Surrey, Guildford, UK
| | - Veni Ezhil
- 1 Royal Surrey County Hospital, University of Surrey, Guildford, UK
| | - Vineet Prakash
- 1 Royal Surrey County Hospital, University of Surrey, Guildford, UK
| | - Sheaka Alobaidli
- 3 Centre for Vision, Speech and Signal Processing, University of Surrey, Guildford, UK
| | | | | | - James Scuffham
- 1 Royal Surrey County Hospital, University of Surrey, Guildford, UK
| | - Andrew Nisbet
- 1 Royal Surrey County Hospital, University of Surrey, Guildford, UK
| | - Philip Evans
- 3 Centre for Vision, Speech and Signal Processing, University of Surrey, Guildford, UK
| |
Collapse
|
236
|
Wang L. Screening and Biosensor-Based Approaches for Lung Cancer Detection. SENSORS (BASEL, SWITZERLAND) 2017; 17:2420. [PMID: 29065541 PMCID: PMC5677261 DOI: 10.3390/s17102420] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/17/2017] [Accepted: 10/18/2017] [Indexed: 02/07/2023]
Abstract
Early diagnosis of lung cancer helps to reduce the cancer death rate significantly. Over the years, investigators worldwide have extensively investigated many screening modalities for lung cancer detection, including computerized tomography, chest X-ray, positron emission tomography, sputum cytology, magnetic resonance imaging and biopsy. However, these techniques are not suitable for patients with other pathologies. Developing a rapid and sensitive technique for early diagnosis of lung cancer is urgently needed. Biosensor-based techniques have been recently recommended as a rapid and cost-effective tool for early diagnosis of lung tumor markers. This paper reviews the recent development in screening and biosensor-based techniques for early lung cancer detection.
Collapse
Affiliation(s)
- Lulu Wang
- School of Instrument Science and Opto-electronics Engineering, Hefei University of Technology, Hefei 230009, China.
- Institute of Biomedical Technologies, Auckland University of Technology, Auckland 1142, New Zealand.
| |
Collapse
|
237
|
Sadeghi-Naini A, Suraweera H, Tran WT, Hadizad F, Bruni G, Rastegar RF, Curpen B, Czarnota GJ. Breast-Lesion Characterization using Textural Features of Quantitative Ultrasound Parametric Maps. Sci Rep 2017; 7:13638. [PMID: 29057899 PMCID: PMC5651882 DOI: 10.1038/s41598-017-13977-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 10/04/2017] [Indexed: 12/19/2022] Open
Abstract
This study evaluated, for the first time, the efficacy of quantitative ultrasound (QUS) spectral parametric maps in conjunction with texture-analysis techniques to differentiate non-invasively benign versus malignant breast lesions. Ultrasound B-mode images and radiofrequency data were acquired from 78 patients with suspicious breast lesions. QUS spectral-analysis techniques were performed on radiofrequency data to generate parametric maps of mid-band fit, spectral slope, spectral intercept, spacing among scatterers, average scatterer diameter, and average acoustic concentration. Texture-analysis techniques were applied to determine imaging biomarkers consisting of mean, contrast, correlation, energy and homogeneity features of parametric maps. These biomarkers were utilized to classify benign versus malignant lesions with leave-one-patient-out cross-validation. Results were compared to histopathology findings from biopsy specimens and radiology reports on MR images to evaluate the accuracy of technique. Among the biomarkers investigated, one mean-value parameter and 14 textural features demonstrated statistically significant differences (p < 0.05) between the two lesion types. A hybrid biomarker developed using a stepwise feature selection method could classify the legions with a sensitivity of 96%, a specificity of 84%, and an AUC of 0.97. Findings from this study pave the way towards adapting novel QUS-based frameworks for breast cancer screening and rapid diagnosis in clinic.
Collapse
Affiliation(s)
- Ali Sadeghi-Naini
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Harini Suraweera
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - William Tyler Tran
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Centre for Health and Social Care Research, Sheffield Hallam University, Sheffield, UK
| | - Farnoosh Hadizad
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Giancarlo Bruni
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | - Belinda Curpen
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Gregory J Czarnota
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada. .,Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. .,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
238
|
Liu G, Huang SY, Franc B, Seo Y, Mitra D. Unsupervised Learning in PET Radiomics. IEEE NUCLEAR SCIENCE SYMPOSIUM CONFERENCE RECORD. NUCLEAR SCIENCE SYMPOSIUM 2017; 2017:10.1109/NSSMIC.2017.8532959. [PMID: 30631241 PMCID: PMC6324844 DOI: 10.1109/nssmic.2017.8532959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In this study, we investigated large scale radoimics on 116 breast cancer patients. We are particularly interested in unsupervised learning to bicluster patients and features in order to associate such biclusters with the disease characteristics. The results show that radiomics features with wavelet features have a better biclustering ability. And 172 radiomics features have shown a better classification capability.
Collapse
Affiliation(s)
- G Liu
- School of Computing, Florida Institute of Technology, Melbourne, FL
| | - S-Y Huang
- Radiology Department, University of California San Francisco
| | - B Franc
- Radiology Department, University of California San Francisco
| | - Y Seo
- Radiology Department, University of California San Francisco
| | - D Mitra
- School of Computing, Florida Institute of Technology, Melbourne, FL
| |
Collapse
|
239
|
Cook GJR, Lovat E, Siddique M, Goh V, Ferner R, Warbey VS. Characterisation of malignant peripheral nerve sheath tumours in neurofibromatosis-1 using heterogeneity analysis of 18F-FDG PET. Eur J Nucl Med Mol Imaging 2017; 44:1845-1852. [PMID: 28589254 PMCID: PMC5644685 DOI: 10.1007/s00259-017-3733-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 05/12/2017] [Indexed: 12/27/2022]
Abstract
PURPOSE Measurement of heterogeneity in 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) images is reported to improve tumour phenotyping and response assessment in a number of cancers. We aimed to determine whether measurements of 18F-FDG heterogeneity could improve differentiation of benign symptomatic neurofibromas from malignant peripheral nerve sheath tumours (MPNSTs). METHODS 18F-FDG PET data from a cohort of 54 patients (24 female, 30 male, mean age 35.1 years) with neurofibromatosis-1 (NF1), and clinically suspected malignant transformation of neurofibromas into MPNSTs, were included. Scans were performed to a standard clinical protocol at 1.5 and 4 h post-injection. Six first-order [including three standardised uptake value (SUV) parameters], four second-order (derived from grey-level co-occurrence matrices) and four high-order (derived from neighbourhood grey-tone difference matrices) statistical features were calculated from tumour volumes of interest. Each patient had histological verification or at least 5 years clinical follow-up as the reference standard with regards to the characterisation of tumours as benign (n = 30) or malignant (n = 24). RESULTS There was a significant difference between benign and malignant tumours for all six first-order parameters (at 1.5 and 4 h; p < 0.0001), for second-order entropy (only at 4 h) and for all high-order features (at 1.5 h and 4 h, except contrast at 4 h; p < 0.0001-0.047). Similarly, the area under the receiver operating characteristic curves was high (0.669-0.997, p < 0.05) for the same features as well as 1.5-h second-order entropy. No first-, second- or high-order feature performed better than maximum SUV (SUVmax) at differentiating benign from malignant tumours. CONCLUSIONS 18F-FDG uptake in MPNSTs is higher than benign symptomatic neurofibromas, as defined by SUV parameters, and more heterogeneous, as defined by first- and high-order heterogeneity parameters. However, heterogeneity analysis does not improve on SUVmax discriminative performance.
Collapse
Affiliation(s)
- Gary J. R. Cook
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
| | - Eitan Lovat
- Guy’s, Kings and St Thomas’ Medical School, King’s College London, London, UK
| | - Muhammad Siddique
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
| | - Vicky Goh
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
| | - Rosalie Ferner
- National Neurofibromatosis Service, Department of Neurology, Guys & St Thomas’ NHS Foundation Trust, London, UK
| | - Victoria S. Warbey
- Cancer Imaging Department, Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, SE1 7EH UK
| |
Collapse
|
240
|
Maley CC, Aktipis A, Graham TA, Sottoriva A, Boddy AM, Janiszewska M, Silva AS, Gerlinger M, Yuan Y, Pienta KJ, Anderson KS, Gatenby R, Swanton C, Posada D, Wu CI, Schiffman JD, Hwang ES, Polyak K, Anderson ARA, Brown JS, Greaves M, Shibata D. Classifying the evolutionary and ecological features of neoplasms. Nat Rev Cancer 2017; 17:605-619. [PMID: 28912577 PMCID: PMC5811185 DOI: 10.1038/nrc.2017.69] [Citation(s) in RCA: 259] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neoplasms change over time through a process of cell-level evolution, driven by genetic and epigenetic alterations. However, the ecology of the microenvironment of a neoplastic cell determines which changes provide adaptive benefits. There is widespread recognition of the importance of these evolutionary and ecological processes in cancer, but to date, no system has been proposed for drawing clinically relevant distinctions between how different tumours are evolving. On the basis of a consensus conference of experts in the fields of cancer evolution and cancer ecology, we propose a framework for classifying tumours that is based on four relevant components. These are the diversity of neoplastic cells (intratumoural heterogeneity) and changes over time in that diversity, which make up an evolutionary index (Evo-index), as well as the hazards to neoplastic cell survival and the resources available to neoplastic cells, which make up an ecological index (Eco-index). We review evidence demonstrating the importance of each of these factors and describe multiple methods that can be used to measure them. Development of this classification system holds promise for enabling clinicians to personalize optimal interventions based on the evolvability of the patient's tumour. The Evo- and Eco-indices provide a common lexicon for communicating about how neoplasms change in response to interventions, with potential implications for clinical trials, personalized medicine and basic cancer research.
Collapse
Affiliation(s)
- Carlo C Maley
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 1001 S. McAllister Ave, Tempe, Arizona 85287, USA
| | - Athena Aktipis
- Department of Psychology, Center for Evolution and Medicine, Arizona State University, 651 E. University Drive, Tempe, Arizona 85287, USA
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Amy M Boddy
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Michalina Janiszewska
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue D740C, Boston, Massachusetts 02215, USA
| | - Ariosto S Silva
- Department of Cancer Imaging and Metabolism, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Marco Gerlinger
- Centre for Evolution and Cancer, Division of Molecular Pathology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Yinyin Yuan
- Centre for Evolution and Cancer, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Kenneth J Pienta
- Brady Urological Institute, The Johns Hopkins School of Medicine, 600 N. Wolfe Street, Baltimore, Maryland 21287, USA
| | - Karen S Anderson
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 1001 S. McAllister Ave, Tempe, Arizona 85287, USA
| | - Robert Gatenby
- Cancer Biology and Evolution Program, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - David Posada
- Department of Biochemistry, Genetics and Immunology and Biomedical Research Center (CINBIO), University of Vigo, Spain; Galicia Sur Health Research Institute, Vigo, 36310, Spain
| | - Chung-I Wu
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois 60637, USA
| | - Joshua D Schiffman
- Departments of Pediatrics and Oncological Sciences, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, Utah 84108, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University and Duke Cancer Institute, 465 Seeley Mudd Building, Durham, North Carolina 27710, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue D740C, Boston, Massachusetts 02215, USA
| | - Alexander R A Anderson
- Integrated Mathematical Oncology Department, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Joel S Brown
- Integrated Mathematical Oncology Department, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Darryl Shibata
- Department of Pathology, Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Avenue, NOR2424, Los Angeles, California 90033, USA
| |
Collapse
|
241
|
Moscoso A, Ruibal Á, Domínguez-Prado I, Fernández-Ferreiro A, Herranz M, Albaina L, Argibay S, Silva-Rodríguez J, Pardo-Montero J, Aguiar P. Texture analysis of high-resolution dedicated breast 18 F-FDG PET images correlates with immunohistochemical factors and subtype of breast cancer. Eur J Nucl Med Mol Imaging 2017; 45:196-206. [DOI: 10.1007/s00259-017-3830-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 09/04/2017] [Indexed: 01/28/2023]
|
242
|
Tsujikawa T, Yamamoto M, Shono K, Yamada S, Tsuyoshi H, Kiyono Y, Kimura H, Okazawa H, Yoshida Y. Assessment of intratumor heterogeneity in mesenchymal uterine tumor by an 18F-FDG PET/CT texture analysis. Ann Nucl Med 2017; 31:752-757. [DOI: 10.1007/s12149-017-1208-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/08/2017] [Indexed: 01/30/2023]
|
243
|
Altazi BA, Zhang GG, Fernandez DC, Montejo ME, Hunt D, Werner J, Biagioli MC, Moros EG. Reproducibility of F18-FDG PET radiomic features for different cervical tumor segmentation methods, gray-level discretization, and reconstruction algorithms. J Appl Clin Med Phys 2017; 18:32-48. [PMID: 28891217 PMCID: PMC5689938 DOI: 10.1002/acm2.12170] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 01/18/2023] Open
Abstract
Site‐specific investigations of the role of radiomics in cancer diagnosis and therapy are emerging. We evaluated the reproducibility of radiomic features extracted from 18Flourine–fluorodeoxyglucose (18F‐FDG) PET images for three parameters: manual versus computer‐aided segmentation methods, gray‐level discretization, and PET image reconstruction algorithms. Our cohort consisted of pretreatment PET/CT scans from 88 cervical cancer patients. Two board‐certified radiation oncologists manually segmented the metabolic tumor volume (MTV1 and MTV2) for each patient. For comparison, we used a graphical‐based method to generate semiautomated segmented volumes (GBSV). To address any perturbations in radiomic feature values, we down‐sampled the tumor volumes into three gray‐levels: 32, 64, and 128 from the original gray‐level of 256. Finally, we analyzed the effect on radiomic features on PET images of eight patients due to four PET 3D‐reconstruction algorithms: maximum likelihood‐ordered subset expectation maximization (OSEM) iterative reconstruction (IR) method, fourier rebinning‐ML‐OSEM (FOREIR), FORE‐filtered back projection (FOREFBP), and 3D‐Reprojection (3DRP) analytical method. We extracted 79 features from all segmentation method, gray‐levels of down‐sampled volumes, and PET reconstruction algorithms. The features were extracted using gray‐level co‐occurrence matrices (GLCM), gray‐level size zone matrices (GLSZM), gray‐level run‐length matrices (GLRLM), neighborhood gray‐tone difference matrices (NGTDM), shape‐based features (SF), and intensity histogram features (IHF). We computed the Dice coefficient between each MTV and GBSV to measure segmentation accuracy. Coefficient values close to one indicate high agreement, and values close to zero indicate low agreement. We evaluated the effect on radiomic features by calculating the mean percentage differences (d¯) between feature values measured from each pair of parameter elements (i.e. segmentation methods: MTV1‐MTV2, MTV1‐GBSV, MTV2‐GBSV; gray‐levels: 64‐32, 64‐128, and 64‐256; reconstruction algorithms: OSEM‐FORE‐OSEM, OSEM‐FOREFBP, and OSEM‐3DRP). We used |d¯| as a measure of radiomic feature reproducibility level, where any feature scored |d¯| ±SD ≤ |25|% ± 35% was considered reproducible. We used Bland–Altman analysis to evaluate the mean, standard deviation (SD), and upper/lower reproducibility limits (U/LRL) for radiomic features in response to variation in each testing parameter. Furthermore, we proposed U/LRL as a method to classify the level of reproducibility: High— ±1% ≤ U/LRL ≤ ±30%; Intermediate— ±30% < U/LRL ≤ ±45%; Low— ±45 < U/LRL ≤ ±50%. We considered any feature below the low level as nonreproducible (NR). Finally, we calculated the interclass correlation coefficient (ICC) to evaluate the reliability of radiomic feature measurements for each parameter. The segmented volumes of 65 patients (81.3%) scored Dice coefficient >0.75 for all three volumes. The result outcomes revealed a tendency of higher radiomic feature reproducibility among segmentation pair MTV1‐GBSV than MTV2‐GBSV, gray‐level pairs of 64‐32 and 64‐128 than 64‐256, and reconstruction algorithm pairs of OSEM‐FOREIR and OSEM‐FOREFBP than OSEM‐3DRP. Although the choice of cervical tumor segmentation method, gray‐level value, and reconstruction algorithm may affect radiomic features, some features were characterized by high reproducibility through all testing parameters. The number of radiomic features that showed insensitivity to variations in segmentation methods, gray‐level discretization, and reconstruction algorithms was 10 (13%), 4 (5%), and 1 (1%), respectively. These results suggest that a careful analysis of the effects of these parameters is essential prior to any radiomics clinical application.
Collapse
Affiliation(s)
- Baderaldeen A Altazi
- Department of Radiation Oncology, H.L. Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Department of Physics, University of South Florida, Tampa, FL, USA.,Department of Radiation Oncology, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Geoffrey G Zhang
- Department of Radiation Oncology, H.L. Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Department of Physics, University of South Florida, Tampa, FL, USA
| | - Daniel C Fernandez
- Department of Radiation Oncology, H.L. Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Michael E Montejo
- Department of Radiation Oncology, H.L. Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Dylan Hunt
- Department of Radiation Oncology, H.L. Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Joan Werner
- Department of Radiation Oncology, H.L. Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Eduardo G Moros
- Department of Radiation Oncology, H.L. Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Department of Physics, University of South Florida, Tampa, FL, USA
| |
Collapse
|
244
|
Textural features and SUV-based variables assessed by dual time point 18F-FDG PET/CT in locally advanced breast cancer. Ann Nucl Med 2017; 31:726-735. [PMID: 28887761 PMCID: PMC5691106 DOI: 10.1007/s12149-017-1203-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 08/30/2017] [Indexed: 11/26/2022]
Abstract
Aim To study the influence of dual time point 18F-FDG PET/CT in textural features and SUV-based variables and their relation among them. Methods Fifty-six patients with locally advanced breast cancer (LABC) were prospectively included. All of them underwent a standard 18F-FDG PET/CT (PET-1) and a delayed acquisition (PET-2). After segmentation, SUV variables (SUVmax, SUVmean, and SUVpeak), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) were obtained. Eighteen three-dimensional (3D) textural measures were computed including: run-length matrices (RLM) features, co-occurrence matrices (CM) features, and energies. Differences between all PET-derived variables obtained in PET-1 and PET-2 were studied. Results Significant differences were found between the SUV-based parameters and MTV obtained in the dual time point PET/CT, with higher values of SUV-based variables and lower MTV in the PET-2 with respect to the PET-1. In relation with the textural parameters obtained in dual time point acquisition, significant differences were found for the short run emphasis, low gray-level run emphasis, short run high gray-level emphasis, run percentage, long run emphasis, gray-level non-uniformity, homogeneity, and dissimilarity. Textural variables showed relations with MTV and TLG. Conclusion Significant differences of textural features were found in dual time point 18F-FDG PET/CT. Thus, a dynamic behavior of metabolic characteristics should be expected, with higher heterogeneity in delayed PET acquisition compared with the standard PET. A greater heterogeneity was found in bigger tumors.
Collapse
|
245
|
Diagnostic importance of 18F-FDG PET/CT parameters and total lesion glycolysis in differentiating between benign and malignant adrenal lesions. Nucl Med Commun 2017; 38:788-794. [DOI: 10.1097/mnm.0000000000000712] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
246
|
Rahmim A, Huang P, Shenkov N, Fotouhi S, Davoodi-Bojd E, Lu L, Mari Z, Soltanian-Zadeh H, Sossi V. Improved prediction of outcome in Parkinson's disease using radiomics analysis of longitudinal DAT SPECT images. Neuroimage Clin 2017; 16:539-544. [PMID: 29868437 PMCID: PMC5984570 DOI: 10.1016/j.nicl.2017.08.021] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/14/2017] [Accepted: 08/24/2017] [Indexed: 02/01/2023]
Abstract
No disease modifying therapies for Parkinson's disease (PD) have been found effective to date. To properly power clinical trials for discovery of such therapies, the ability to predict outcome in PD is critical, and there is a significant need for discovery of prognostic biomarkers of PD. Dopamine transporter (DAT) SPECT imaging is widely used for diagnostic purposes in PD. In the present work, we aimed to evaluate whether longitudinal DAT SPECT imaging can significantly improve prediction of outcome in PD patients. In particular, we investigated whether radiomics analysis of DAT SPECT images, in addition to use of conventional non-imaging and imaging measures, could be used to predict motor severity at year 4 in PD subjects. We selected 64 PD subjects (38 male, 26 female; age at baseline (year 0): 61.9 ± 7.3, range [46,78]) from the Parkinson's Progressive Marker Initiative (PPMI) database. Inclusion criteria included (i) having had at least 2 SPECT scans at years 0 and 1 acquired on a similar scanner, (ii) having undergone a high-resolution 3 T MRI scan, and (iii) having motor assessment (MDS-UPDRS-III) available in year 4 used as outcome measure. Image analysis included automatic region-of-interest (ROI) extraction on MRI images, registration of SPECT images onto the corresponding MRI images, and extraction of radiomic features. Non-imaging predictors included demographics, disease duration as well as motor and non-motor clinical measures in years 0 and 1. The image predictors included 92 radiomic features extracted from the caudate, putamen, and ventral striatum of DAT SPECT images at years 0 and 1 to quantify heterogeneity and texture in uptake. Random forest (RF) analysis with 5000 trees was used to combine both non-imaging and imaging variables to predict motor outcome (UPDRS-III: 27.3 ± 14.7, range [3,77]). The RF prediction was evaluated using leave-one-out cross-validation. Our results demonstrated that addition of radiomic features to conventional measures significantly improved (p < 0.001) prediction of outcome, reducing the absolute error of predicting MDS-UPDRS-III from 9.00 ± 0.88 to 4.12 ± 0.43. This shows that radiomics analysis of DAT SPECT images has a significant potential towards development of effective prognostic biomarkers in PD.
Collapse
Affiliation(s)
- Arman Rahmim
- Department of Radiology, Johns Hopkins University, Baltimore, United States
- Department of Electrical & Computer Engineering, Johns Hopkins University, Baltimore, United States
| | - Peng Huang
- Departments of Oncology and Biostatistics, Johns Hopkins University, Baltimore, United States
| | - Nikolay Shenkov
- Department of Physics & Astronomy, University of British Columbia, Vancouver, Canada
| | - Sima Fotouhi
- Department of Radiology, Johns Hopkins University, Baltimore, United States
| | - Esmaeil Davoodi-Bojd
- Departments of Radiology and Research Administration, Henry Ford Health System, Detroit, MI, United States
| | - Lijun Lu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Zoltan Mari
- Department of Neurology and Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
| | - Hamid Soltanian-Zadeh
- Departments of Radiology and Research Administration, Henry Ford Health System, Detroit, MI, United States
- CIPCE, School of Electrical & Computer Engineering, University of Tehran, Tehran, Iran
| | - Vesna Sossi
- Department of Physics & Astronomy, University of British Columbia, Vancouver, Canada
| |
Collapse
|
247
|
18F-FDG PET radiomics approaches: comparing and clustering features in cervical cancer. Ann Nucl Med 2017; 31:678-685. [DOI: 10.1007/s12149-017-1199-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/05/2017] [Indexed: 12/13/2022]
|
248
|
Comparison of image quality between step-and-shoot and continuous bed motion techniques in whole-body 18F-fluorodeoxyglucose positron emission tomography with the same acquisition duration. Ann Nucl Med 2017; 31:686-695. [PMID: 28815414 DOI: 10.1007/s12149-017-1200-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/06/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVE This study aimed to compare the qualities of whole-body positron emission tomography (PET) images acquired by the step-and-shoot (SS) and continuous bed motion (CBM) techniques with approximately the same acquisition duration, through phantom and clinical studies. METHODS A body phantom with 10-37 mm spheres was filled with 18F-fluorodeoxyglucose (FDG) solution at a sphere-to-background radioactivity ratio of 4:1 and acquired by both techniques. Reconstructed images were evaluated by visual assessment, percentages of contrast (%Q H) and background variability (%N) in accordance with the Japanese guideline for oncology FDG-PET/computed tomography (CT). To evaluate the variability of the standardized uptake value (SUV), the coefficient of variation (CV) for both maximum SUV and peak SUV was examined. Both the SUV values were additionally compared with those of standard images acquired for 30 min, and their accuracy was evaluated by the %difference (%Diff). In the clinical study, whole-body 18F-FDG PET/CT images of 60 patients acquired by both techniques were compared for liver signal-to-noise ratio (SNRliver), CV at end planes, and both SUV values. RESULTS In the phantom study, the visual assessment and %Q H values of the two techniques did not differ from each other. However, the %N values of the CBM technique were significantly higher than those of the SS technique. Additionally, the CV and %Diff for both SUV values in the CBM images tended to be slightly higher than those in SS images. In the clinical study, the SNRliver values of CBM images were significantly lower than those of SS images, although the CV at the end planes in CBM images was significantly lower than those in SS images. In the Bland-Altman analysis for both SUV values, the mean differences were close to 0, and most lesions exhibited SUVs within the limits of agreement. CONCLUSIONS The CBM technique exhibited slightly lesser uniformity in the center plane than the SS technique. Additionally, in the phantom study, the CV and %Diff of SUV values in CBM images tended to be slightly higher than those of SS images. However, since these differences were subtle, they might be negligible in clinical settings.
Collapse
|
249
|
Ben Bouallègue F, Tabaa YA, Kafrouni M, Cartron G, Vauchot F, Mariano-Goulart D. Association between textural and morphological tumor indices on baseline PET-CT and early metabolic response on interim PET-CT in bulky malignant lymphomas. Med Phys 2017; 44:4608-4619. [DOI: 10.1002/mp.12349] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Fayçal Ben Bouallègue
- Nuclear Medicine Department; Gui de Chauliac University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
| | - Yassine Al Tabaa
- Nuclear Medicine Department; Gui de Chauliac University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
| | - Marilyne Kafrouni
- Nuclear Medicine Department; Gui de Chauliac University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
| | - Guillaume Cartron
- Haematology Department; Saint Eloi University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
| | - Fabien Vauchot
- Nuclear Medicine Department; Gui de Chauliac University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
| | - Denis Mariano-Goulart
- Nuclear Medicine Department; Gui de Chauliac University Hospital; 80, Avenue Augustin Fliche 34295 Montpellier Cedex 5 France
- U1046 INSERM - UMR9214 CNRS; CHU Arnaud de Villeneuve; 371 Avenue du Doyen Giraud 34295 Montpellier Cedex 5 France
| |
Collapse
|
250
|
Zhai TT, van Dijk LV, Huang BT, Lin ZX, Ribeiro CO, Brouwer CL, Oosting SF, Halmos GB, Witjes MJH, Langendijk JA, Steenbakkers RJHM, Sijtsema NM. Improving the prediction of overall survival for head and neck cancer patients using image biomarkers in combination with clinical parameters. Radiother Oncol 2017; 124:256-262. [PMID: 28764926 DOI: 10.1016/j.radonc.2017.07.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/25/2017] [Accepted: 07/13/2017] [Indexed: 02/05/2023]
Abstract
PURPOSE To develop and validate prediction models of overall survival (OS) for head and neck cancer (HNC) patients based on image biomarkers (IBMs) of the primary tumor and positive lymph nodes (Ln) in combination with clinical parameters. MATERIAL AND METHODS The study cohort was composed of 289 nasopharyngeal cancer (NPC) patients from China and 298 HNC patients from the Netherlands. Multivariable Cox-regression analysis was performed to select clinical parameters from the NPC and HNC datasets, and IBMs from the NPC dataset. Final prediction models were based on both IBMs and clinical parameters. RESULTS Multivariable Cox-regression analysis identified three independent IBMs (tumor Volume-density, Run Length Non-uniformity and Ln Major-axis-length). This IBM model showed a concordance(c)-index of 0.72 (95%CI: 0.65-0.79) for the NPC dataset, which performed reasonably with a c-index of 0.67 (95%CI: 0.62-0.72) in the external validation HNC dataset. When IBMs were added in clinical models, the c-index of the NPC and HNC datasets improved to 0.75 (95%CI: 0.68-0.82; p=0.019) and 0.75 (95%CI: 0.70-0.81; p<0.001), respectively. CONCLUSION The addition of IBMs from the primary tumor and Ln improved the prognostic performance of the models containing clinical factors only. These combined models may improve pre-treatment individualized prediction of OS for HNC patients.
Collapse
Affiliation(s)
- Tian-Tian Zhai
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands; Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, China.
| | - Lisanne V van Dijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Bao-Tian Huang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, China
| | - Zhi-Xiong Lin
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, China.
| | - Cássia O Ribeiro
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Charlotte L Brouwer
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Sjoukje F Oosting
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Gyorgy B Halmos
- Department of Otolaryngology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Max J H Witjes
- Department of Maxillofacial Surgery, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Johannes A Langendijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Roel J H M Steenbakkers
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Nanna M Sijtsema
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| |
Collapse
|