1
|
Chirra PV, Giriprakash P, Rizk AG, Kurowski JA, Viswanath SE, Gandhi NS. Developing a Reproducible Radiomics Model for Diagnosis of Active Crohn's Disease on CT Enterography Across Annotation Variations and Acquisition Differences. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025; 38:1594-1605. [PMID: 39466507 DOI: 10.1007/s10278-024-01303-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024]
Abstract
To systematically identify radiomics features on CT enterography (CTE) scans which can accurately diagnose active Crohn's disease across multiple sources of variation. Retrospective study of CTE scans curated between 2013 and 2015, comprising 164 subjects (65 male, 99 female; all patients were over the age of 18) with endoscopic confirmation for the presence or absence of active Crohn's disease. All patients had three distinct sets of scans available (full and reduced dose, where the latter had been reconstructed via two different methods), acquired on a single scanner at a single institution. Radiomics descriptors from annotated terminal ileum regions were individually and systematically evaluated for resilience to different imaging variations (changes in dose/reconstruction, batch effects, and simulated annotation differences) via multiple reproducibility measures. Multiple radiomics models (by accounting for each source of variation) were evaluated in terms of classifier area under the ROC curve (AUC) for identifying patients with active Crohn's disease, across separate discovery and hold-out validation cohorts. Radiomics descriptors selected based on resiliency to multiple sources of imaging variation yielded the highest overall classification performance in the discovery cohort (AUC = 0.79 ± 0.04) which also best generalized in hold-out validation (AUC = 0.81). Performance was maintained across multiple doses and reconstructions while also being significantly better (p < 0.001) than non-resilient descriptors or descriptors only resilient to a single source of variation. Radiomics features can accurately diagnose active Crohn's disease on CTE scans across multiple sources of imaging variation via systematic analysis of reproducibility measures. Clinical utility and translatability of radiomics features for diagnosis and characterization of Crohn's disease on CTE scans will be contingent on their reproducibility across multiple types and sources of imaging variation.
Collapse
Affiliation(s)
- Prathyush V Chirra
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Pavithran Giriprakash
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Alain G Rizk
- Section, Abdominal Imaging, Imaging Institute, and Digestive Diseases and Surgery Institute and Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jacob A Kurowski
- Department of Pediatric Gastroenterology, Hepatology & Nutrition, Cleveland Clinic, Cleveland, OH, USA
| | - Satish E Viswanath
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
- Cleveland Veterns Affairs Medical Center, Cleveland, OH, USA.
| | - Namita S Gandhi
- Section, Abdominal Imaging, Imaging Institute, and Digestive Diseases and Surgery Institute and Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
2
|
Yang SX, Li M, Zhou LN, Hou DH, Zhang L, Wu N. Reproducibility of the CT radiomic features of pulmonary nodules: the effects of the CT reconstruction algorithm, radiation dose, and contrast agent. Quant Imaging Med Surg 2025; 15:2309-2318. [PMID: 40160618 PMCID: PMC11948441 DOI: 10.21037/qims-24-2026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 01/14/2025] [Indexed: 04/02/2025]
Abstract
Background The reproducibility of radiomic features (RFs) is essential in lung nodule diagnosis. This study aimed to prospectively investigate the effects of computed tomography (CT) scanning parameters on the reproducibility of RFs in pulmonary nodules. Methods Patients with pulmonary nodules who underwent chest CT scans at the Cancer Hospital of the Chinese Academy of Medical Sciences between July 2018 and March 2019 were prospectively included in the study. Six sequences with three pairs of different scanning parameters, including the reconstruction algorithm [filtered back projection (FBP) vs. 50% adaptive statistical iterative reconstruction-V (ASiR-V)], radiation dose (low dose vs. standard dose), and contrast agent [contrast-enhanced (CE) CT vs. non-contrast enhanced (NE) CT], were used for each patient. When one of the scanning parameters was changed, the other two remained fixed. The nodules were classified into pure ground-glass nodules (pGGNs), part-solid nodules (PSNs), and solid nodules (SNs) according to the nodule consistency. RFs with an intraclass correlation coefficient (ICC) >0.75 were considered to have good retest reliability. All the RF values of the different scanning parameters and nodule consistency were investigated and compared. Results A total of 150 pulmonary nodules, including 50 pGGNs, 50 PSNs, and 50 SNs, in 96 patients (mean age: 52±10 years; 62 females) were included in the study. In total, 320 RFs with an ICC >0.75 were evaluated. The proportion of RFs showed significant difference between FBP and 50% ASiR-V, low dose and standard dose, and CE and NE CT scans was 38.4% (123/320), 63.1% (202/320) and 54.1% (173/320), respectively. The radiation dose and contrast agent affected more RFs than the reconstruction algorithm (both P<0.001). In the subgroup analysis of nodule consistency, regardless of changes in the reconstruction algorithms, radiation doses, or contrast agents, the RFs showed significant difference among the pGGNs, PSNs, and SNs (all P<0.001). Conclusions The scanning parameters affected the reproducibility of the RFs, and nodules of different consistency were affected differently. The effects of these parameters should be fully considered in radiomic analysis.
Collapse
Affiliation(s)
- Shou-Xin Yang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Meng Li
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Na Zhou
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong-Hui Hou
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Zhang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Wu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Nuclear Medicine (PET-CT Center), National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Zhao B, Dercle L, Yang H, Riely GJ, Kris MG, Schwartz LH. Annotated test-retest dataset of lung cancer CT scan images reconstructed at multiple imaging parameters. Sci Data 2024; 11:1259. [PMID: 39567508 PMCID: PMC11579286 DOI: 10.1038/s41597-024-04085-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
Quantitative imaging biomarkers (QIB) are increasingly used in clinical research to advance precision medicine approaches in oncology. Computed tomography (CT) is a modality of choice for cancer diagnosis, prognosis, and response assessment due to its reliability and global accessibility. Here, we contribute to the cancer imaging community through The Cancer Imaging Archive (TCIA) by providing investigator-initiated, same-day repeat CT scan images of 32 non-small cell lung cancer (NSCLC) patients, along with radiologist-annotated lesion contours as a reference standard. Each scan was reconstructed into 6 image settings using various combinations of three slice thicknesses (1.25 mm, 2.5 mm, 5 mm) and two reconstruction kernels (lung, standard; GE CT equipment), which spans a wide range of CT imaging reconstruction parameters commonly used in lung cancer clinical practice and clinical trials. This holds considerable value for advancing the development of robust Radiomics, Artificial Intelligence (AI) and machine learning (ML) methods.
Collapse
Affiliation(s)
- Binsheng Zhao
- Memorial Sloan-Kettering Cancer Center, New York, NY, 10021, USA.
| | - Laurent Dercle
- Memorial Sloan-Kettering Cancer Center, New York, NY, 10021, USA
- Department of Radiology, Columbia University New York, New York, NY, 10032, USA
| | - Hao Yang
- Memorial Sloan-Kettering Cancer Center, New York, NY, 10021, USA
| | - Gregory J Riely
- Memorial Sloan-Kettering Cancer Center, New York, NY, 10021, USA
| | - Mark G Kris
- Memorial Sloan-Kettering Cancer Center, New York, NY, 10021, USA
| | | |
Collapse
|
4
|
Jeong J, Park D, Kang JH, Kim M, Kim HY, Choi W, Ham SY. Deep Learning-Based Slice Thickness Reduction for Computer-Aided Detection of Lung Nodules in Thick-Slice CT. Diagnostics (Basel) 2024; 14:2558. [PMID: 39594224 PMCID: PMC11592715 DOI: 10.3390/diagnostics14222558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Computer-aided detection (CAD) systems for lung nodule detection often face challenges with 5 mm computed tomography (CT) scans, leading to missed nodules. This study assessed the efficacy of a deep learning-based slice thickness reduction technique from 5 mm to 1 mm to enhance CAD performance. METHODS In this retrospective study, 687 chest CT scans were analyzed, including 355 with nodules and 332 without nodules. CAD performance was evaluated on nodules, to which all three radiologists agreed. RESULTS The slice thickness reduction technique significantly improved the area under the receiver operating characteristic curve (AUC) for scan-level analysis from 0.867 to 0.902, with a p-value < 0.001, and nodule-level sensitivity from 0.826 to 0.916 at two false positives per scan. Notably, the performance showed greater improvements on smaller nodules than larger nodules. Qualitative analysis confirmed that nodules mistaken for ground glass on 5 mm scans could be correctly identified as part-solid on the refined 1 mm CT, thereby improving the diagnostic capability. CONCLUSIONS Applying a deep learning-based slice thickness reduction technique significantly enhances CAD performance in lung nodule detection on chest CT scans, supporting the clinical adoption of refined 1 mm CT scans for more accurate diagnoses.
Collapse
Affiliation(s)
- Jonghun Jeong
- VUNO Inc., Seoul 06541, Republic of Korea; (J.J.); (D.P.); (J.-H.K.)
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Doohyun Park
- VUNO Inc., Seoul 06541, Republic of Korea; (J.J.); (D.P.); (J.-H.K.)
| | - Jung-Hyun Kang
- VUNO Inc., Seoul 06541, Republic of Korea; (J.J.); (D.P.); (J.-H.K.)
| | - Myungsub Kim
- Department of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea; (M.K.); (W.C.)
| | - Hwa-Young Kim
- Department of Radiology, CHA Gangnam Medical Center, CHA University, Seoul 06125, Republic of Korea;
| | - Woosuk Choi
- Department of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea; (M.K.); (W.C.)
| | - Soo-Youn Ham
- Department of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Republic of Korea; (M.K.); (W.C.)
| |
Collapse
|
5
|
Xie J, He Y, Che S, Zhao W, Niu Y, Qin D, Li Z. Differential diagnosis of benign and lung adenocarcinoma presenting as larger solid nodules and masses based on multiscale CT radiomics. PLoS One 2024; 19:e0309033. [PMID: 39365772 PMCID: PMC11451992 DOI: 10.1371/journal.pone.0309033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/04/2024] [Indexed: 10/06/2024] Open
Abstract
PURPOSE To develop a better radiomic model for the differential diagnosis of benign and lung adenocarcinoma lesions presenting as larger solid nodules and masses based on multiscale computed tomography (CT) radiomics. MATERIALS AND METHODS This retrospective study enrolled 205 patients with solid nodules and masses from Center 1 between January 2010 and February 2022 and Center 2 between January 2019 and February 2022. After applying the inclusion and exclusion criteria, we retrospectively enrolled 165 patients from two centers and assigned them to the training dataset (n = 115) or the test dataset (n = 50). Radiomics features were extracted from volumes of interest on CT images. A gradient boosting decision tree (GBDT) was used for data dimensionality reduction to perform the final feature selection. Four models were developed using clinical data, conventional imaging features and radiomics features, namely, the clinical and image model (CIM), the plain CT radiomics model (PRM), the enhanced CT radiomics model (ERM) and the combined model (CM). Model performance was evaluated to determine the best model for identifying benign and lung adenocarcinoma presenting as larger solid nodules and masses. RESULTS In the training dataset, the areas under the curve (AUCs) for the CIM, PRM, ERM, and CM were 0.718, 0.806, 0.819, and 0.917, respectively. The differential diagnostic capability of the ERM was better than that of the PRM and the CIM. The CM was optimal. Intermediate and junior radiologists and respiratory physicians achieved improved obviously diagnostic results with the radiomics model. The senior radiologists showed slight improved diagnostic results after using the radiomics model. CONCLUSION Radiomics may have the potential to be used as a noninvasive tool for the differential diagnosis of benign and lung adenocarcinoma lesions presenting as larger solid nodules and masses.
Collapse
Affiliation(s)
- Jiayue Xie
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Yifan He
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Siyu Che
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Wenjing Zhao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Yuxin Niu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Dongxue Qin
- Department of Radiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Zhiyong Li
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
6
|
Coppes RP, van Dijk LV. Future of Team-based Basic and Translational Science in Radiation Oncology. Semin Radiat Oncol 2024; 34:370-378. [PMID: 39271272 DOI: 10.1016/j.semradonc.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
To further optimize radiotherapy, a more personalized treatment towards individual patient's risk profiles, dissecting both patient-specific tumor and normal tissue response to multimodality treatments is needed. Novel developments in radiobiology, using in vitro patient-specific complex tissue resembling 3D models and multiomics approaches at a spatial single-cell level, may provide unprecedented insight into the radiation responses of tumors and normal tissue. Here, we describe the necessary team effort, including all disciplines in radiation oncology, to integrate such data into clinical prediction models and link the relatively "big data" from the clinical practice, allowing accurate patient stratification for personalized treatment approaches.
Collapse
Affiliation(s)
- R P Coppes
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.; Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, Groningen, Netherlands..
| | - L V van Dijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
7
|
Tran K, Ginzburg D, Hong W, Attenberger U, Ko HS. Post-radiotherapy stage III/IV non-small cell lung cancer radiomics research: a systematic review and comparison of CLEAR and RQS frameworks. Eur Radiol 2024; 34:6527-6543. [PMID: 38625613 PMCID: PMC11399214 DOI: 10.1007/s00330-024-10736-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/07/2024] [Accepted: 03/04/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Lung cancer, the second most common cancer, presents persistently dismal prognoses. Radiomics, a promising field, aims to provide novel imaging biomarkers to improve outcomes. However, clinical translation faces reproducibility challenges, despite efforts to address them with quality scoring tools. OBJECTIVE This study had two objectives: 1) identify radiomics biomarkers in post-radiotherapy stage III/IV nonsmall cell lung cancer (NSCLC) patients, 2) evaluate research quality using the CLEAR (CheckList_for_EvaluAtion_of_Radiomics_research), RQS (Radiomics_Quality_Score) frameworks, and formulate an amalgamated CLEAR-RQS tool to enhance scientific rigor. MATERIALS AND METHODS A systematic literature review (Jun-Aug 2023, MEDLINE/PubMed/SCOPUS) was conducted concerning stage III/IV NSCLC, radiotherapy, and radiomic features (RF). Extracted data included study design particulars, such as sample size, radiotherapy/CT technique, selected RFs, and endpoints. CLEAR and RQS were merged into a CLEAR-RQS checklist. Three readers appraised articles utilizing CLEAR, RQS, and CLEAR-RQS metrics. RESULTS Out of 871 articles, 11 met the inclusion/exclusion criteria. The Median cohort size was 91 (range: 10-337) with 9 studies being single-center. No common RF were identified. The merged CLEAR-RQS checklist comprised 61 items. Most unreported items were within CLEAR's "methods" and "open-source," and within RQS's "phantom-calibration," "registry-enrolled prospective-trial-design," and "cost-effective-analysis" sections. No study scored above 50% on RQS. Median CLEAR scores were 55.74% (32.33/58 points), and for RQS, 17.59% (6.3/36 points). CLEAR-RQS article ranking fell between CLEAR and RQS and aligned with CLEAR. CONCLUSION Radiomics research in post-radiotherapy stage III/IV NSCLC exhibits variability and frequently low-quality reporting. The formulated CLEAR-RQS checklist may facilitate education and holds promise for enhancing radiomics research quality. CLINICAL RELEVANCE STATEMENT Current radiomics research in the field of stage III/IV postradiotherapy NSCLC is heterogenous, lacking reproducibility, with no identified imaging biomarker. Radiomics research quality assessment tools may enhance scientific rigor and thereby facilitate radiomics translation into clinical practice. KEY POINTS There is heterogenous and low radiomics research quality in postradiotherapy stage III/IV nonsmall cell lung cancer. Barriers to reproducibility are small cohort size, nonvalidated studies, missing technical parameters, and lack of data, code, and model sharing. CLEAR (CheckList_for_EvaluAtion_of_Radiomics_research), RQS (Radiomics_Quality_Score), and the amalgamated CLEAR-RQS tool are useful frameworks for assessing radiomics research quality and may provide a valuable resource for educational purposes in the field of radiomics.
Collapse
Affiliation(s)
- Kevin Tran
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, VIC 3052, Australia
| | - Daniel Ginzburg
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany
| | - Wei Hong
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Ulrike Attenberger
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany
| | - Hyun Soo Ko
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany.
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, 305 Grattan St, Melbourne, VIC 3000, Australia.
| |
Collapse
|
8
|
Ibrahim A, Guha S, Lu L, Geng P, Wu Q, Chou Y, Yang H, Wang D, Schwartz LH, Xie CM, Zhao B. The reproducibility and predictivity of radiomic features extracted from dynamic contrast-enhanced computed tomography of hepatocellular carcinoma. PLoS One 2024; 19:e0310486. [PMID: 39269960 PMCID: PMC11398651 DOI: 10.1371/journal.pone.0310486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
PURPOSE To assess the reproducibility of radiomic features (RFs) extracted from dynamic contrast-enhanced computed tomography (DCE-CT) scans of patients diagnosed with hepatocellular carcinoma (HCC) with regards to inter-observer variability and acquisition timing after contrast injection. The predictive ability of reproducible RFs for differentiating between the degrees of HCC differentiation is also investigated. METHODS We analyzed a set of DCE-CT scans of 39 patients diagnosed with HCC. Two radiologists independently segmented the scans, and RFs were extracted from each sequence of the DCE-CT scans. The same lesion was segmented across the DCE-CT sequences of each patient's scan. From each lesion, 127 commonly used RFs were extracted. The reproducibility of RFs was assessed with regard to (i) inter-observer variability, by evaluating the reproducibility of RFs between the two radiologists; and (ii) timing of acquisition following contrast injection (inter- and intra-imaging phase). The reproducibility of RFs was assessed using the concordance correlation coefficient (CCC), with a cut-off value of 0.90. Reproducible RFs were used for building XGBoost classification models for the differentiation of HCC differentiation. RESULTS Inter-observer analyses across the different contrast-enhancement phases showed that the number of reproducible RFs was 29 (22.8%), 52 (40.9%), and 36 (28.3%) for the non-contrast enhanced, late arterial, and portal venous phases, respectively. Intra- and inter-sequence analyses revealed that the number of reproducible RFs ranged between 1 (0.8%) and 47 (37%), inversely related with time interval between the sequences. XGBoost algorithms built using reproducible RFs in each phase were found to be high predictive ability of the degree of HCC tumor differentiation. CONCLUSIONS The reproducibility of many RFs was significantly impacted by inter-observer variability, and a larger number of RFs were impacted by the difference in the time of acquisition after contrast injection. Our findings highlight the need for quality assessment to ensure that scans are analyzed in the same physiologic imaging phase in quantitative imaging studies, or that phase-wide reproducible RFs are selected. Overall, the study emphasizes the importance of reproducibility and quality control when using RFs as biomarkers for clinical applications.
Collapse
Affiliation(s)
- Abdalla Ibrahim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Siddharth Guha
- Department of Radiology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Lin Lu
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Pengfei Geng
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Qian Wu
- First Affiliated Hospital of Nanjing Medical University, Jiangsu, China
| | - Yen Chou
- Department of Medical Imaging, Fu Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Hao Yang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Delin Wang
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Lawrence H. Schwartz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Chuan-miao Xie
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Binsheng Zhao
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| |
Collapse
|
9
|
Kalasauskas D, Kosterhon M, Kurz E, Schmidt L, Altmann S, Grauhan NF, Sommer C, Othman A, Brockmann MA, Ringel F, Keric N. Preoperative prediction of CNS WHO grade and tumour aggressiveness in intracranial meningioma based on radiomics and structured semantics. Sci Rep 2024; 14:20586. [PMID: 39232068 PMCID: PMC11374997 DOI: 10.1038/s41598-024-71200-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024] Open
Abstract
Preoperative identification of intracranial meningiomas with aggressive behaviour may help in choosing the optimal treatment strategy. Radiomics is emerging as a powerful diagnostic tool with potential applications in patient risk stratification. In this study, we aimed to compare the predictive value of conventional, semantic based and radiomic analyses to determine CNS WHO grade and early tumour relapse in intracranial meningiomas. We performed a single-centre retrospective analysis of intracranial meningiomas operated between 2007 and 2018. Recurrence within 5 years after Simpson Grade I-III resection was considered as early. Preoperative T1 CE MRI sequences were analysed conventionally by two radiologists. Additionally a semantic feature score based on systematic analysis of morphological characteristics was developed and a radiomic analysis were performed. For the radiomic model, tumour volume was extracted manually, 791 radiomic features were extracted. Eight feature selection algorithms and eight machine learning methods were used. Models were analysed using test and training datasets. In total, 226 patients were included. There were 21% CNS WHO grade 2 tumours, no CNS WHO grade 3 tumour, and 25 (11%) tumour recurrences were detected in total. In ROC analysis the best radiomic models demonstrated superior performance for determination of CNS WHO grade (AUC 0.930) and early recurrence (AUC 0.892) in comparison to the semantic feature score (AUC 0.74 and AUC 0.65) and conventional radiological analysis (AUC 0.65 and 0.54). The combination of human classifiers, semantic score and radiomic analysis did not markedly increase the model performance. Radiomic analysis is a promising tool for preoperative identification of aggressive and atypical intracranial meningiomas and could become a useful tool in the future.
Collapse
Affiliation(s)
- Darius Kalasauskas
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Michael Kosterhon
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Elena Kurz
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Leon Schmidt
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Sebastian Altmann
- Department of Neuroradiology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Nils F Grauhan
- Department of Neuroradiology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Clemens Sommer
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Ahmed Othman
- Department of Neuroradiology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Marc A Brockmann
- Department of Neuroradiology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Florian Ringel
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Naureen Keric
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| |
Collapse
|
10
|
Badesha AS, Frood R, Bailey MA, Coughlin PM, Scarsbrook AF. A Scoping Review of Machine-Learning Derived Radiomic Analysis of CT and PET Imaging to Investigate Atherosclerotic Cardiovascular Disease. Tomography 2024; 10:1455-1487. [PMID: 39330754 PMCID: PMC11435603 DOI: 10.3390/tomography10090108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Cardiovascular disease affects the carotid arteries, coronary arteries, aorta and the peripheral arteries. Radiomics involves the extraction of quantitative data from imaging features that are imperceptible to the eye. Radiomics analysis in cardiovascular disease has largely focused on CT and MRI modalities. This scoping review aims to summarise the existing literature on radiomic analysis techniques in cardiovascular disease. METHODS MEDLINE and Embase databases were searched for eligible studies evaluating radiomic techniques in living human subjects derived from CT, MRI or PET imaging investigating atherosclerotic disease. Data on study population, imaging characteristics and radiomics methodology were extracted. RESULTS Twenty-nine studies consisting of 5753 patients (3752 males) were identified, and 78.7% of patients were from coronary artery studies. Twenty-seven studies employed CT imaging (19 CT carotid angiography and 6 CT coronary angiography (CTCA)), and two studies studied PET/CT. Manual segmentation was most frequently undertaken. Processing techniques included voxel discretisation, voxel resampling and filtration. Various shape, first-order, second-order and higher-order radiomic features were extracted. Logistic regression was most commonly used for machine learning. CONCLUSION Most published evidence was feasibility/proof of concept work. There was significant heterogeneity in image acquisition, segmentation techniques, processing and analysis between studies. There is a need for the implementation of standardised imaging acquisition protocols, adherence to published reporting guidelines and economic evaluation.
Collapse
Affiliation(s)
- Arshpreet Singh Badesha
- Department of Radiology, St. James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
| | - Russell Frood
- Department of Radiology, St. James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
- Faculty of Medicine and Health, University of Leeds, Leeds LS2 9TJ, UK
| | - Marc A. Bailey
- Faculty of Medicine and Health, University of Leeds, Leeds LS2 9TJ, UK
- The Leeds Vascular Institute, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK
| | - Patrick M. Coughlin
- The Leeds Vascular Institute, Leeds General Infirmary, Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK
| | - Andrew F. Scarsbrook
- Department of Radiology, St. James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
- Faculty of Medicine and Health, University of Leeds, Leeds LS2 9TJ, UK
| |
Collapse
|
11
|
Bobholz SA, Lowman AK, Connelly JM, Duenweg SR, Winiarz A, Nath B, Kyereme F, Brehler M, Bukowy J, Coss D, Lupo JM, Phillips JJ, Ellingson BM, Krucoff MO, Mueller WM, Banerjee A, LaViolette PS. Noninvasive Autopsy-Validated Tumor Probability Maps Identify Glioma Invasion Beyond Contrast Enhancement. Neurosurgery 2024; 95:537-547. [PMID: 38501824 PMCID: PMC11302944 DOI: 10.1227/neu.0000000000002898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/09/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND AND OBJECTIVES This study identified a clinically significant subset of patients with glioma with tumor outside of contrast enhancement present at autopsy and subsequently developed a method for detecting nonenhancing tumor using radio-pathomic mapping. We tested the hypothesis that autopsy-based radio-pathomic tumor probability maps would be able to noninvasively identify areas of infiltrative tumor beyond traditional imaging signatures. METHODS A total of 159 tissue samples from 65 subjects were aligned to MRI acquired nearest to death for this retrospective study. Demographic and survival characteristics for patients with and without tumor beyond the contrast-enhancing margin were computed. An ensemble algorithm was used to predict pixelwise tumor presence from pathological annotations using segmented cellularity (Cell), extracellular fluid, and cytoplasm density as input (6 train/3 test subjects). A second level of ensemble algorithms was used to predict voxelwise Cell, extracellular fluid, and cytoplasm on the full data set (43 train/22 test subjects) using 5-by-5 voxel tiles from T1, T1 + C, fluid-attenuated inversion recovery, and apparent diffusion coefficient as input. The models were then combined to generate noninvasive whole brain maps of tumor probability. RESULTS Tumor outside of contrast was identified in 41.5% of patients, who showed worse survival outcomes (hazard ratio = 3.90, P < .001). Tumor probability maps reliably tracked nonenhancing tumor on a range of local and external unseen data, identifying tumor outside of contrast in 69% of presurgical cases that also showed reduced survival outcomes (hazard ratio = 1.67, P = .027). CONCLUSION This study developed a multistage model for mapping gliomas using autopsy tissue samples as ground truth, which was able to identify regions of tumor beyond traditional imaging signatures.
Collapse
Affiliation(s)
- Samuel A. Bobholz
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Allison K. Lowman
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jennifer M. Connelly
- Department of Neurology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Savannah R. Duenweg
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Aleksandra Winiarz
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Biprojit Nath
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Fitzgerald Kyereme
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michael Brehler
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John Bukowy
- Department of Electrical Engineering and Computer Science, Milwaukee School of Engineering, Milwaukee, Wisconsin, USA
| | - Dylan Coss
- Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Janine M. Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
- UCSF/UC Berkeley Graduate Program in Bioengineering, University of California, San Francisco and Berkeley, California, USA
| | - Joanna J. Phillips
- Department of Neurological Surgery, University of California, San Francisco, California, USA
- Department of Pathology, University of California, San Francisco, California, USA
| | - Benjamin M. Ellingson
- UCLA Brain Tumor Imaging Laboratory, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Max O. Krucoff
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Wade M. Mueller
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Anjishnu Banerjee
- Department of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Peter S. LaViolette
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
12
|
Song X, Li L, Yu Q, Liu N, Zhu S, Yuan S. Radiogenomics models for predicting prognosis in locally advanced non-small cell lung cancer patients undergoing definitive chemoradiotherapy. Transl Lung Cancer Res 2024; 13:1828-1840. [PMID: 39263037 PMCID: PMC11384488 DOI: 10.21037/tlcr-24-145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/17/2024] [Indexed: 09/13/2024]
Abstract
Background Definitive chemoradiotherapy (dCRT) is the cornerstone for locally advanced non-small cell lung cancer (LA-NSCLC). The study aimed to construct a multi-omics model integrating baseline clinical data, computed tomography (CT) images and genetic information to predict the prognosis of dCRT in LA-NSCLC patients. Methods The study retrospectively enrolled 105 stage III LA-NSCLC patients who had undergone dCRT. The pre-treatment CT images were collected, and the primary tumor was delineated as a region of interest (ROI) on the image using 3D-Slicer, and the radiomics features were extracted. The least absolute shrinkage and selection operator (LASSO) was employed for dimensionality reduction and selection of features. Genomic information was obtained from the baseline tumor tissue samples. We then constructed a multi-omics model by combining baseline clinical data, radiomics and genomics features. The predictive performance of the model was evaluated by the area under the curve (AUC) of the receiver operating characteristic (ROC) and the concordance index (C-index). Results The median follow-up time was 30.1 months, and the median progression-free survival (PFS) was 10.60 months. Four features were applied to construct the radiomics model. Multivariable analysis demonstrated the Rad-score, KEAP1 and MET mutations were independent prognostic factors for PFS. The C-index of radiomics model, genomics model and radiogenomics model all performed well in the training group (0.590 vs. 0.606 vs. 0.663) and the validation group (0.599 vs. 0.594 vs. 0.650). Conclusions The radiomics model, genomics model and radiogenomics model can all predict the prognosis of dCRT for LA-NSCLC, and the radiogenomics model is superior to the single type model.
Collapse
Affiliation(s)
- Xiaoyu Song
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Li Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, Hefei, China
| | - Qingxi Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ning Liu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, Hefei, China
| | - Shouhui Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shuanghu Yuan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, Hefei, China
| |
Collapse
|
13
|
Yang F, Feng Y, Sun P, Traverso A, Dekker A, Zhang B, Huang Z, Wang Z, Yan D. Preoperative prediction of high-grade osteosarcoma response to neoadjuvant therapy based on a plain CT radiomics model: A dual-center study. J Bone Oncol 2024; 47:100614. [PMID: 38975332 PMCID: PMC11225658 DOI: 10.1016/j.jbo.2024.100614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/05/2024] [Accepted: 06/01/2024] [Indexed: 07/09/2024] Open
Abstract
Objective To develop a model combining clinical and radiomics features from CT scans for a preoperative noninvasive evaluation of Huvos grading of neoadjuvant chemotherapy in patients with HOS. Methods 183 patients from center A and 42 from center B were categorized into training and validation sets. Features derived from radiomics were obtained from unenhanced CT scans.Following dimensionality reduction, the most optimal features were selected and utilized in creating a radiomics model through logistic regression analysis. Integrating clinical features, a composite clinical radiomics model was developed, and a nomogram was constructed. Predictive performance of the model was evaluated using ROC curves and calibration curves. Additionally, decision curve analysis was conducted to assess practical utility of nomogram in clinical settings. Results LASSO LR analysis was performed, and finally, three selected image omics features were obtained.Radiomics model yielded AUC values with a good diagnostic effect for both patient sets (AUCs: 0.69 and 0.68, respectively). Clinical models (including sex, age, pre-chemotherapy ALP and LDH levels, new lung metastases within 1 year after surgery, and incidence) performed well in terms of Huvos grade prediction, with an AUC of 0.74 for training set. The AUC for independent validation set stood at 0.70. Notably, the amalgamation of radiomics and clinical features exhibited commendable predictive prowess in training set, registering an AUC of 0.78. This robust performance was subsequently validated in the independent validation set, where the AUC remained high at 0.75. Calibration curves of nomogram showed that the predictions were in good agreement with actual observations. Conclusion Combined model can be used for Huvos grading in patients with HOS after preoperative chemotherapy, which is helpful for adjuvant treatment decisions.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation, Beijing Jishuitan Hospital,Capital Medical University, Beijing 100035, China
| | - Ying Feng
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Pengfei Sun
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Alberto Traverso
- Department of Radiation Oncology (Maastro), GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Bin Zhang
- Department of Radiation, Peking University Shougang Hospital, Beijing 100144, China
| | - Zhen Huang
- Department of Bone Oncology, Beijing Jishuitan Hospital,Capital Medical University, Beijing 100035, China
| | - Zhixiang Wang
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Dong Yan
- Department of Radiation, Beijing Jishuitan Hospital,Capital Medical University, Beijing 100035, China
| |
Collapse
|
14
|
Brown KH, Kerr BN, Pettigrew M, Connor K, Miller IS, Shiels L, Connolly C, McGarry C, Byrne AT, Butterworth KT. A comparative analysis of preclinical computed tomography radiomics using cone-beam and micro-computed tomography scanners. Phys Imaging Radiat Oncol 2024; 31:100615. [PMID: 39157293 PMCID: PMC11328005 DOI: 10.1016/j.phro.2024.100615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/17/2024] [Accepted: 07/20/2024] [Indexed: 08/20/2024] Open
Abstract
Background and purpose Radiomics analysis extracts quantitative data (features) from medical images. These features could potentially reflect biological characteristics and act as imaging biomarkers within precision medicine. However, there is a lack of cross-comparison and validation of radiomics outputs which is paramount for clinical implementation. In this study, we compared radiomics outputs across two computed tomography (CT)-based preclinical scanners. Materials and methods Cone beam CT (CBCT) and µCT scans were acquired using different preclinical CT imaging platforms. The reproducibility of radiomics features on each scanner was assessed using a phantom across imaging energies (40 & 60 kVp) and segmentation volumes (44-238 mm3). Retrospective mouse scans were used to compare feature reliability across varying tissue densities (lung, heart, bone), scanners and after voxel size harmonisation. Reliable features had an intraclass correlation coefficient (ICC) > 0.8. Results First order and GLCM features were the most reliable on both scanners across different volumes. There was an inverse relationship between tissue density and feature reliability, with the highest number of features in lung (CBCT=580, µCT=734) and lowest in bone (CBCT=110, µCT=560). Comparable features for lung and heart tissues increased when voxel sizes were harmonised. We have identified tissue-specific preclinical radiomics signatures in mice for the lung (133), heart (35), and bone (15). Conclusions Preclinical CBCT and µCT scans can be used for radiomics analysis to support the development of meaningful radiomics signatures. This study demonstrates the importance of standardisation and emphasises the need for multi-centre studies.
Collapse
Affiliation(s)
- Kathryn H Brown
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Brianna N Kerr
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Mihaela Pettigrew
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Kate Connor
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Ian S Miller
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- National Preclinical Imaging Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Liam Shiels
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Colum Connolly
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Conor McGarry
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
- Northern Ireland Cancer Centre, Belfast Health & Social Care Trust, Belfast, United Kingdom
| | - Annette T Byrne
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- National Preclinical Imaging Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Karl T Butterworth
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
15
|
Zheng J, Hao Y, Guo Y, Du M, Wang P, Xin J. An 18F-FDG-PET/CT-based radiomics signature for estimating malignance probability of solitary pulmonary nodule. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e13751. [PMID: 38725315 PMCID: PMC11082539 DOI: 10.1111/crj.13751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/29/2024] [Accepted: 03/28/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Some solitary pulmonary nodules (SPNs) as early manifestations of lung cancer, it is difficult to determine its nature, which brings great trouble to clinical diagnosis and treatment. Radiomics can deeply explore the essence of images and provide clinical decision support for clinicians. The purpose of our study was to explore the effect of positron emission tomography (PET) with 2-deoxy-2-[fluorine-18] fluoro-d-glucose integrated with computed tomography (CT; 18F-FDG-PET/CT) combined with radiomics for predicting probability of malignancy of SPNs. METHODS We retrospectively enrolled 190 patients with SPNs confirmed by pathology from January 2013 to December 2019 in our hospital. SPNs were benign in 69 patients and malignant in 121 patients. Patients were randomly divided into a training or testing group at a ratio of 7:3. Three-dimensional regions of interest (ROIs) were manually outlined on PET and CT images, and radiomics features were extracted. Synthetic minority oversampling technique (SMOTE) method was used to balance benign and malignant samples to a ratio of 1:1. In the training group, least absolute shrinkage and selection operator (LASSO) regression analyses and Spearman correlation analyses were used to select the strongest radiomics features. Three models including PET model, CT model, and joint model were constructed using multivariate logistic regression analysis. Receiver operating characteristic (ROC) curves, calibration curves, and decision curves were plotted to evaluate diagnostic efficiency, calibration degree, and clinical usefulness of all models in training and testing groups. RESULTS The estimative effectiveness of the joint model was superior to the CT or PET model alone in the training and testing groups. For the joint model, CT model, and PET model, area under the ROC curve was 0.929, 0.819, 0.833 in the training group, and 0.844, 0.759, 0.748 in the testing group, respectively. Calibration and decision curves showed good fit and clinical usefulness for the joint model in both training and testing groups. CONCLUSION Radiomics models constructed by combining PET and CT radiomics features are valuable for distinguishing benign and malignant SPNs. The combined effect is superior to qualitative diagnoses with CT or PET radiomics models alone.
Collapse
Affiliation(s)
- Jingchi Zheng
- Radiology DepartmentShengjing Hospital of China Medical UniversityShenyangChina
| | - Yue Hao
- Radiology DepartmentShengjing Hospital of China Medical UniversityShenyangChina
| | | | - Ming Du
- Nuclear Medicine DepartmentShengjing Hospital of China Medical UniversityShenyangChina
| | - Pengyuan Wang
- Nuclear Medicine DepartmentShengjing Hospital of China Medical UniversityShenyangChina
| | - Jun Xin
- Nuclear Medicine DepartmentShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
16
|
Huang Y, Chen L, Ding Q, Zhang H, Zhong Y, Zhang X, Weng S. CT-based radiomics for predicting pathological grade in hepatocellular carcinoma. Front Oncol 2024; 14:1295575. [PMID: 38690170 PMCID: PMC11059035 DOI: 10.3389/fonc.2024.1295575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 03/18/2024] [Indexed: 05/02/2024] Open
Abstract
Objective To construct and validate radiomics models for hepatocellular carcinoma (HCC) grade predictions based on contrast-enhanced CT (CECT). Methods Patients with pathologically confirmed HCC after surgery and underwent CECT at our institution between January 2016 and December 2020 were enrolled and randomly divided into training and validation datasets. With tumor segmentation and feature extraction, radiomic models were constructed using univariate analysis, followed by least absolute shrinkage and selection operator (LASSO) regression. In addition, combined models with clinical factors and radiomics scores (Radscore) were constructed using logistic regression. Finally, all models were evaluated using the receiver operating characteristic (ROC) curve with the area under the curve (AUC), calibration curve, and decision curve analysis (DCA). Results In total 242 patients were enrolled in this study, of whom 170 and 72 formed the training and validation datasets, respectively. The arterial phase and portal venous phase (AP+VP) radiomics model were evaluated as the best for predicting HCC pathological grade among all the models built in our study (AUC = 0.981 in the training dataset; AUC = 0.842 in the validation dataset) and was used to build a nomogram. Furthermore, the calibration curve and DCA indicated that the AP+VP radiomics model had a satisfactory prediction efficiency. Conclusions Low- and high-grade HCC can be distinguished with good diagnostic performance using a CECT-based radiomics model.
Collapse
Affiliation(s)
- Yue Huang
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lingfeng Chen
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qingzhu Ding
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Han Zhang
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yun Zhong
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiang Zhang
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shangeng Weng
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
17
|
Cao J, Li Q, Zhang H, Wu Y, Wang X, Ding S, Chen S, Xu S, Duan G, Qiu D, Sun J, Shi J, Liu S. Radiomics model based on MRI to differentiate spinal multiple myeloma from metastases: A two-center study. J Bone Oncol 2024; 45:100599. [PMID: 38601920 PMCID: PMC11004638 DOI: 10.1016/j.jbo.2024.100599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 12/19/2023] [Accepted: 01/09/2024] [Indexed: 04/12/2024] Open
Abstract
Purpose Spinal multiple myeloma (MM) and metastases are two common cancer types with similar imaging characteristics, for which differential diagnosis is needed to ensure precision therapy. The aim of this study is to establish radiomics models for effective differentiation between them. Methods Enrolled in this study were 263 patients from two medical institutions, including 127 with spinal MM and 136 with spinal metastases. Of them, 210 patients from institution I were used as the internal training cohort and 53 patients from Institution II were used as the external validation cohort. Contrast-enhanced T1-weighted imaging (CET1) and T2-weighted imaging (T2WI) sequences were collected and reviewed. Based on the 1037 radiomics features extracted from both CET1 and T2WI images, Logistic Regression (LR), AdaBoost (AB), Support Vector Machines (SVM), Random Forest (RF), and multiple kernel learning based SVM (MKL-SVM) were constructed. Hyper-parameters were tuned by five-fold cross-validation. The diagnostic efficiency among different radiomics models was compared by accuracy (ACC), sensitivity (SEN), specificity (SPE), area under the ROC curve (AUC), YI, positive predictive value (PPV), negative predictive value (NPY), and F1-score. Results Based on single-sequence, the RF model outperformed all other models. All models based on T2WI images performed better than those based on CET1. The efficiency of all models was boosted by incorporating CET1 and T2WI sequences, and the MKL-SVM model achieved the best performance with ACC, AUC, and F1-score of 0.862, 0.870, and 0.874, respectively. Conclusions The radiomics models constructed based on MRI achieved satisfactory diagnostic performance for differentiation of spinal MM and metastases, demonstrating broad application prospects for individualized diagnosis and treatment.
Collapse
Affiliation(s)
- Jiashi Cao
- Department of Orthopedics, Navy Medical Center, the Navy Medical University, No. 338 Huaihai West Road, Shanghai 200052, China
| | - Qiong Li
- Department of Radiology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center/Cancer Hospital, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Huili Zhang
- School of Communication and Information Engineering, Shanghai University, 99 Shangda Road, Baoshan District, Shanghai 200444, China
| | - Yanyan Wu
- Department of Radiology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Xiang Wang
- Department of Radiology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Saisai Ding
- School of Communication and Information Engineering, Shanghai University, 99 Shangda Road, Baoshan District, Shanghai 200444, China
| | - Song Chen
- Department of Radiology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Shaochun Xu
- Department of Radiology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Guangwen Duan
- Department of Radiology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| | - Defu Qiu
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou 221116, China
| | - Jiuyi Sun
- Department of Orthopedics, Navy Medical Center, the Navy Medical University, No. 338 Huaihai West Road, Shanghai 200052, China
| | - Jun Shi
- School of Communication and Information Engineering, Shanghai University, 99 Shangda Road, Baoshan District, Shanghai 200444, China
| | - Shiyuan Liu
- Department of Radiology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai 200003, China
| |
Collapse
|
18
|
Wang TW, Chao HS, Chiu HY, Lu CF, Liao CY, Lee Y, Chen JR, Shiao TH, Chen YM, Wu YT. Radiomics of metastatic brain tumor as a predictive image biomarker of progression-free survival in patients with non-small-cell lung cancer with brain metastasis receiving tyrosine kinase inhibitors. Transl Oncol 2024; 39:101826. [PMID: 37984256 PMCID: PMC10689936 DOI: 10.1016/j.tranon.2023.101826] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 10/18/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Epidermal growth factor receptor (EGFR)-targeted tyrosine kinase inhibitors (TKIs) are the first-line therapy for EGFR-mutant non-small-cell lung cancer (NSCLC). Early prediction of treatment failure in patients with brain metastases treated with EGFR-TKIs may help in making decisions for systemic drug therapy or local brain tumor control. This study examined the predictive power of the radiomics of both brain metastasis tumors and primary lung tumors. We propose a deep learning based CoxCC model based on quantitative brain magnetic resonance imaging (MRI), a prognostic index and clinical data; the model can be used to predict progression-free survival (PFS) after EGFR-TKI therapy in advanced EGFR-mutant NSCLC. METHODS This retrospective single-center study included 271 patients receiving first-line EGFR-TKI targeted therapy in 2018-2019. Among them, 72 patients who had brain metastases before receiving first-line EGFR-TKI treatment. Three radiomic features were extracted from pretreatment brain MRI images. A CoxCC model for the progression risk stratification of EGFR-TKI treatment was proposed on the basis of MRI radiomics, clinical features, and a prognostic index. We performed time-dependent PFS predictions to evaluate the performance of the CoxCC model. RESULTS The CoxCC model based on a prognostic index, clinical features, and radiomic features of brain metastasis exhibited higher performance than clinical features combined with indexes previously proposed for determining the prognosis of brain metastasis, including recursive partitioning analysis, diagnostic-specific graded prognostic assessment, graded prognostic assessment for lung cancer using molecular markers (lung-molGPA), and modified lung-molGPA, with c-index values of 0.75, 0.67, 0.66, 0.65, and 0.65, respectively. The model achieved areas under the curve of 0.88, 0.73, 0.92, and 0.90 for predicting PFS at 3, 6, 9 and 12 months, respectively. PFS significantly differed between the high- and low-risk groups (p < 0.001). CONCLUSIONS For patients with advanced-stage NSCLC with brain metastasis, MRI radiomics of brain metastases may predict PFS. The CoxCC model integrating brain metastasis radiomics, clinical features, and a prognostic index provided reliable multi-time-point PFS predictions for patients with advanced NSCLC and brain metastases receiving EGFR-TKI treatment.
Collapse
Affiliation(s)
- Ting-Wei Wang
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Heng-Sheng Chao
- Department of Chest Medicine, Taipei Veteran General Hospital, Taipei, Taiwan
| | - Hwa-Yen Chiu
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Chest Medicine, Taipei Veteran General Hospital, Taipei, Taiwan
| | - Chia-Feng Lu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chien-Yi Liao
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen Lee
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jyun-Ru Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tsu-Hui Shiao
- Department of Chest Medicine, Taipei Veteran General Hospital, Taipei, Taiwan
| | - Yuh-Min Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Chest Medicine, Taipei Veteran General Hospital, Taipei, Taiwan
| | - Yu-Te Wu
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
19
|
Zhang X, Liu B, Liu K, Wang L. The diagnosis performance of convolutional neural network in the detection of pulmonary nodules: a systematic review and meta-analysis. Acta Radiol 2023; 64:2987-2998. [PMID: 37743663 DOI: 10.1177/02841851231201514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
BACKGROUND Pulmonary nodules are an early imaging indication of lung cancer, and early detection of pulmonary nodules can improve the prognosis of lung cancer. As one of the applications of machine learning, the convolutional neural network (CNN) applied to computed tomography (CT) imaging data improves the accuracy of diagnosis, but the results could be more consistent. PURPOSE To evaluate the diagnostic performance of CNN in assisting in detecting pulmonary nodules in CT images. MATERIAL AND METHODS PubMed, Cochrane Library, Web of Science, Elsevier, CNKI and Wanfang databases were systematically retrieved before 30 April 2023. Two reviewers searched and checked the full text of articles that might meet the criteria. The reference criteria are joint diagnoses by experienced physicians. The pooled sensitivity, specificity and the area under the summary receiver operating characteristic curve (AUC) were calculated by a random-effects model. Meta-regression analysis was performed to explore potential sources of heterogeneity. RESULTS Twenty-six studies were included in this meta-analysis, involving 2,391,702 regions of interest, comprising segmented images with a few wide pixels. The combined sensitivity and specificity values of the CNN model in detecting pulmonary nodules were 0.93 and 0.95, respectively. The pooled diagnostic odds ratio was 291. The AUC was 0.98. There was heterogeneity in sensitivity and specificity among the studies. The results suggested that data sources, pretreatment methods, reconstruction slice thickness, population source and locality might contribute to the heterogeneity of these eligible studies. CONCLUSION The CNN model can be a valuable diagnostic tool with high accuracy in detecting pulmonary nodules.
Collapse
Affiliation(s)
- Xinyue Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, Department of Epidemiology & Biostatistics, School of Public Health, Southeast University, Nanjing, China
| | - Bo Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, Department of Epidemiology & Biostatistics, School of Public Health, Southeast University, Nanjing, China
| | - Kefu Liu
- Department of radiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Lina Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, Department of Epidemiology & Biostatistics, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
20
|
Shi L, Sheng M, Wei Z, Liu L, Zhao J. CT-Based Radiomics Predicts the Malignancy of Pulmonary Nodules: A Systematic Review and Meta-Analysis. Acad Radiol 2023; 30:3064-3075. [PMID: 37385850 DOI: 10.1016/j.acra.2023.05.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 07/01/2023]
Abstract
RATIONALE AND OBJECTIVES More pulmonary nodules (PNs) have been detected with the wide application of computed tomography (CT) in lung cancer screening. Radiomics is a noninvasive approach to predict the malignancy of PNs. We aimed to systematically evaluate the methodological quality of the eligible studies regarding CT-based radiomics models in predicting the malignancy of PNs and evaluate the model performance of the available studies. MATERIALS AND METHODS PubMed, Embase, and Web of Science were searched to retrieve relevant studies. The methodological quality of the included studies was assessed using the Radiomics Quality Score (RQS) and Prediction model Risk of Bias Assessment Tool. A meta-analysis was conducted to evaluate the performance of CT-based radiomics model. Meta-regression and subgroup analyses were employed to investigate the source of heterogeneity. RESULTS In total, 49 studies were eligible for qualitative analysis and 27 studies were included in quantitative synthesis. The median RQS of 49 studies was 13 (range -2 to 20). The overall risk of bias was found to be high, and the overall applicability was of low concern in all included studies. The pooled sensitivity, specificity, and diagnostic odds ratio were 0.86 95% confidence interval (CI): 0.79-0.91, 0.84 95% CI: 0.78-0.88, and 31.55 95% CI: 21.31-46.70, respectively. The overall area under the curve was 0.91 95% CI: 0.89-0.94. Meta-regression showed the type of PNs on heterogeneity. CT-based radiomics models performed better in studies including only solid PNs. CONCLUSION CT-based radiomics models exhibited excellent diagnostic performance in predicting the malignancy of PNs. Prospective, large sample size, and well-devised studies are desired to verify the prediction capabilities of CT-based radiomics model.
Collapse
Affiliation(s)
- Lili Shi
- Medical School, Nantong University, Nantong, China (L.S., Z.W.)
| | - Meihong Sheng
- Department of Radiology, The Second Affiliated Hospital of Nantong University and Nantong First People's Hospital, Nantong, China (M.S.)
| | - Zhichao Wei
- Medical School, Nantong University, Nantong, China (L.S., Z.W.)
| | - Lei Liu
- Institutes of Intelligence Medicine, Fudan University, Shanghai, China (L.L.)
| | - Jinli Zhao
- Department of Radiology, Affiliated Hospital of Nantong University, Nantong, China (J.Z.).
| |
Collapse
|
21
|
Gupta S, Nayak K, Pendem S. Impact of slice thickness on reproducibility of CT radiomic features of lung tumors. F1000Res 2023; 12:1319. [PMID: 38454921 PMCID: PMC10918310 DOI: 10.12688/f1000research.141148.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 03/09/2024] Open
Abstract
Background Radiomics posits that quantified characteristics from radiographic images reflect underlying pathophysiology. Lung cancer (LC) is one of the prevalent forms of cancer, causing mortality. Slice thickness (ST) of computed tomography (CT) images is a crucial factor influencing the generalizability of radiomic features (RF) in oncology. There is scarcity of research that how ST affects variability of RF in LC. The present study helps in identifying the specific RF categories affected by variations in ST and provides valuable insights for researchers and clinicians working with RF in the field of LC.Hence, aim of the study is to evaluate influence of ST on reproducibility of CT-RF for lung tumors. Methods This is a prospective study, 32 patients with confirmed histopathological diagnosis of lung tumors were included. Contrast Enhanced CT (CECT) thorax was performed using a 128- Incisive CT (Philips Health Care). The image acquisition was performed with 5-mm and 2 mm STwas reconstructed retrospectively. RF were extracted from the CECT thorax images of both ST. We conducted a paired t-test to evaluate the disparity in RF between the two thicknesses. Lin's Concordance Correlation Coefficient (CCC) was performed to identify the reproducibility of RF between the two thicknesses. Results Out of 107 RF, 66 (61.6%) exhibited a statistically significant distinction (p<0.05) when comparing two ST and while 41 (38.3%) RF did not show significant distinction (p>0.05) between the two ST measurements. 29 features (CCC ≥ 0.90) showed excellent to moderate reproducibility, and 78 features (CCC ≤ 0.90) showed poor reproducibility. Among the 7 RF categories, the shape-based features (57.1%) showed the maximum reproducibility whereas NGTDM-based features showed negligible reproducibility. Conclusions The ST had a notable impact on the majority of CT-RF of lung tumors. Shape based features (57.1%). First order (44.4%) features showed highest reproducibility compared to other RF categories.
Collapse
Affiliation(s)
- Sanat Gupta
- Manipal College of Health Professions, Department of Medical Imaging Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kaushik Nayak
- Manipal College of Health Professions, Department of Medical Imaging Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Saikiran Pendem
- Manipal College of Health Professions, Department of Medical Imaging Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
22
|
Sun H, Zhang C, Ouyang A, Dai Z, Song P, Yao J. Multi-classification model incorporating radiomics and clinic-radiological features for predicting invasiveness and differentiation of pulmonary adenocarcinoma nodules. Biomed Eng Online 2023; 22:112. [PMID: 38037082 PMCID: PMC10687925 DOI: 10.1186/s12938-023-01180-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023] Open
Abstract
PURPOSE To develop a comprehensive multi-classification model that combines radiomics and clinic-radiological features to accurately predict the invasiveness and differentiation of pulmonary adenocarcinoma nodules. METHODS A retrospective analysis was conducted on a cohort comprising 500 patients diagnosed with lung adenocarcinoma between January 2020 and December 2022. The dataset included preoperative CT images and histological reports of adenocarcinoma in situ (AIS, n = 97), minimally invasive adenocarcinoma (MIA, n = 139), and invasive adenocarcinoma (IAC, n = 264) with well-differentiated (WIAC, n = 99), moderately differentiated (MIAC, n = 84), and poorly differentiated IAC (PIAC, n = 81). The patients were classified into two groups (IAC and non-IAC) for binary classification and further divided into three and five groups for multi-classification. Feature selection was performed using the least absolute shrinkage and selection operator (LASSO) algorithm to identify the most informative radiomics and clinic-radiological features. Eight machine learning (ML) models were developed using these features, and their performance was evaluated using accuracy (ACC) and the area under the receiver-operating characteristic curve (AUC). RESULTS The combined model, utilizing the support vector machine (SVM) algorithm, demonstrated improved performance in the testing cohort, achieving an AUC of 0.942 and an ACC of 0.894 for the two-classification task. For the three- and five-classification tasks, the combined model employing the one versus one strategy of SVM (SVM-OVO) outperformed other models, with ACC values of 0.767 and 0.607, respectively. The AUC values for histological subtypes ranged from 0.787 to 0.929 in the testing cohort, while the Macro-AUC and Micro-AUC of the multi-classification models ranged from 0.858 to 0.896. CONCLUSIONS A multi-classification radiomics model combined with clinic-radiological features, using the SVM-OVO algorithm, holds promise for accurately predicting the histological characteristics of pulmonary adenocarcinoma nodules, which contributes to personalized treatment strategies for patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Haitao Sun
- Medical Imaging Center, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong Province, China
| | - Chunling Zhang
- Medical Imaging Center, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong Province, China
| | - Aimei Ouyang
- Medical Imaging Center, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong Province, China
| | - Zhengjun Dai
- Scientific Research Department of Huiying Medical Technology Co., Ltd, 66 Xixiaokou Road, Haidian District, Beijing, 100192, China
| | - Peiji Song
- Medical Imaging Center, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong Province, China
| | - Jian Yao
- Medical Imaging Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Huaiyin District, Jinan, 250021, Shandong Province, China.
| |
Collapse
|
23
|
Tang X, Li Y, Shen LT, Yan WF, Qian WL, Yang ZG. CT Radiomics Predict EGFR-T790M Resistance Mutation in Advanced Non-Small Cell Lung Cancer Patients After Progression on First-line EGFR-TKI. Acad Radiol 2023; 30:2574-2587. [PMID: 36941156 DOI: 10.1016/j.acra.2023.01.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 03/23/2023]
Abstract
RATIONALE AND OBJECTIVES We aim to explore the value of chest CT radiomics in predicting the epidermal growth factor receptor (EGFR)-T790M resistance mutation of advanced non-small cell lung cancer (NSCLC) patients after the failure of first-line EGFR-tyrosine kinase inhibitor (EGFR-TKI). MATERIALS AND METHODS A total of 211 and 135 advanced NSCLC patients with tumor tissue-based (Cohort-1) or circulating tumor DNA (ctDNA)-based (Cohort-2) EGFR-T790M testing were included, respectively. Cohort-1 was used for modeling and Cohort-2 was for models' validation. Radiomic features were extracted from tumor lesions on chest nonenhanced CT (NECT) and/or contrast-enhanced CT (CECT). We used eight feature selectors and eight classifier algorithms to establish radiomic models. Models were evaluated by area under the receiver operating characteristic curve (AUC), calibration curve, and decision curve analysis (DCA). RESULTS CT morphological manifestations of peripheral location and pleural indentation sign were associated with EGFR-T790M. For NECT, CECT, and NECT+CECT radiomic features, the feature selector and classifier algorithms of LASSO and Stepwise logistic regression, Boruta and SVM, and LASSO and SVM were chosen to develop the optimal model, respectively (AUC: 0.844, 0.811, and 0.897). All models performed well in calibration curves and DCA. Independent validation of models in Cohort-2 revealed that both NECT and CECT models individually had limited power for predicting EGFR-T790M mutation detected by ctDNA (AUC: 0.649, 0.675), while the NECT+CECT radiomic model had a satisfactory AUC (0.760). CONCLUSION This study proved the feasibility of using CT radiomic features to predict the EGFR-T790M resistance mutation, which could be helpful in guiding personalized therapeutic strategies.
Collapse
Affiliation(s)
- Xin Tang
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Li
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li-Ting Shen
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei-Feng Yan
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wen-Lei Qian
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhi-Gang Yang
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
24
|
Wang TW, Chao HS, Chiu HY, Lin YH, Chen HC, Lu CF, Liao CY, Lee Y, Shiao TH, Chen YM, Huang JW, Wu YT. Evaluating the Potential of Delta Radiomics for Assessing Tyrosine Kinase Inhibitor Treatment Response in Non-Small Cell Lung Cancer Patients. Cancers (Basel) 2023; 15:5125. [PMID: 37958300 PMCID: PMC10647242 DOI: 10.3390/cancers15215125] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Our study aimed to harness the power of CT scans, observed over time, in predicting how lung adenocarcinoma patients might respond to a treatment known as EGFR-TKI. Analyzing scans from 322 advanced stage lung cancer patients, we identified distinct image-based patterns. By integrating these patterns with comprehensive clinical information, such as gene mutations and treatment regimens, our predictive capabilities were significantly enhanced. Interestingly, the precision of these predictions, particularly related to radiomics features, diminished when data from various centers were combined, suggesting that the approach requires standardization across facilities. This novel method offers a potential pathway to anticipate disease progression in lung adenocarcinoma patients treated with EGFR-TKI, laying the groundwork for more personalized treatments. To further validate this approach, extensive studies involving a larger cohort are pivotal.
Collapse
Affiliation(s)
- Ting-Wei Wang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Heng-Sheng Chao
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Hwa-Yen Chiu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Yi-Hui Lin
- Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Hung-Chun Chen
- Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Chia-Feng Lu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chien-Yi Liao
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yen Lee
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Tsu-Hui Shiao
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Yuh-Min Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Jing-Wen Huang
- Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Yu-Te Wu
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
25
|
Malcolm JA, Tacey M, Gibbs P, Lee B, Ko HS. Current state of radiomic research in pancreatic cancer: focusing on study design and reproducibility of findings. Eur Radiol 2023; 33:6659-6669. [PMID: 37079029 PMCID: PMC10511615 DOI: 10.1007/s00330-023-09653-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 04/21/2023]
Abstract
OBJECTIVES To critically appraise methodology and reproducibility of published studies on CT radiomics of pancreatic ductal adenocarcinoma (PDAC). METHODS PRISMA literature search of MEDLINE, PubMed, and Scopus databases was conducted from June to August 2022 relating to CT radiomics human research articles pertaining to PDAC diagnosis, treatment, and/ or prognosis, utilising Image Biomarker Standardisation Initiative-compliant (IBSI) radiomic software. Keyword search included [pancreas OR pancreatic] AND [radiomic OR [quantitative AND imaging] OR [texture AND analysis]]. Analysis included cohort size, CT protocol used, radiomic feature (RF) extraction, segmentation, and selection, software used, outcome correlation, and statistical methodology, with focus on reproducibility. RESULTS Initial search yielded 1112 articles; however, only 12 articles met all inclusion/exclusion criteria. Cohort sizes ranged from 37 to 352 (median = 106, mean = 155.8). CT slice thickness varied among studies (4 using ≤ 1 mm, 5 using > 1 to 3 mm, 2 using > 3 to 5 mm, 1 not specifying). CT protocol varied (5 using a single portal-venous (pv)-phase, 5 using a pancreas protocol, 1 study using a non-contrast protocol). RF extraction and segmentation were heterogeneous (RF extraction: 5 using pv-phase, 2 using late arterial, 4 using multi-phase, 1 using non-contrast phase; RF selection: 3 pre-selected, 9 software-selected). 2D/3D RF segmentation was diverse (2D in 6, 3D in 4, 2D and 3D in 2 studies). Six different radiomics software were used. Research questions and cohort characteristics varied, ultimately leading to non-comparable outcome results. CONCLUSION The current twelve published IBSI-compliant PDAC radiomic studies show high variability and often incomplete methodology resulting in low robustness and reproducibility. CLINICAL RELEVANCE STATEMENT Radiomics research requires IBSI compliance, data harmonisation, and reproducible feature extraction methods for non-invasive imaging biomarker discoveries to be valid. This will ensure a successful clinical implementation and ultimately an improvement of patient outcomes as part of precision and personalised medicine. KEY POINTS • Current state of radiomics research in pancreatic cancer shows low software compliance to the Image Biomarker Standardisation Initiative (IBSI). • IBSI-compliant radiomics studies in pancreatic cancer are heterogeneous and not comparable, and the majority of study designs showed low reproducibility. • Improved methodology and standardisation of practice in the emerging field of radiomics has the potential of this non-invasive imaging biomarker in the management of pancreatic cancer.
Collapse
Affiliation(s)
- James Alex Malcolm
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
- Department of Cancer Imaging, The Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mark Tacey
- Department of Biostatistics, Northern Health, Epping, VIC, Australia
| | - Peter Gibbs
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Oncology, Western Health, Melbourne, VIC, Australia
- Department of Medical Oncology, Melbourne Health, Melbourne, VIC, Australia
| | - Belinda Lee
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Oncology, Melbourne Health, Melbourne, VIC, Australia
- Department of Medical Oncology, The Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Medical Oncology, Northern Health, Epping, VIC, Australia
| | - Hyun Soo Ko
- Department of Cancer Imaging, The Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia.
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
26
|
Li X, Chen J, Zhang C, Han Z, Zheng X, Cao D. Application value of CT radiomic nomogram in predicting T790M mutation of lung adenocarcinoma. BMC Pulm Med 2023; 23:339. [PMID: 37697337 PMCID: PMC10494384 DOI: 10.1186/s12890-023-02609-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/21/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND The purpose of this study was to develop a radiomic nomogram to predict T790M mutation of lung adenocarcinoma base on non-enhanced CT lung images. METHODS This retrospective study reviewed demographic data and lung CT images of 215 lung adenocarcinoma patients with T790M gene test results. 215 patients (including 52 positive) were divided into a training set (n = 150, 36 positive) and an independent test set (n = 65, 16 positive). Multivariate logistic regression was used to select demographic data and CT semantic features to build clinical model. We extracted quantitative features from the volume of interest (VOI) of the lesion, and developed the radiomic model with different feature selection algorithms and classifiers. The models were trained by a 5-fold cross validation strategy on the training set and assessed on the test set. ROC was used to estimate the performance of the clinical model, radiomic model, and merged nomogram. RESULTS Three demographic features (gender, smoking, emphysema) and ten radiomic features (Kruskal-Wallis as selection algorithm, LASSO Logistic Regression as classifier) were determined to build the models. The AUC of the clinical model, radiomic model, and nomogram in the test set were 0.742(95%CI, 0.619-0.843), 0.810(95%CI, 0.696-0.907), 0.841(95%CI, 0.743-0.938), respectively. The predictive efficacy of the nomogram was better than the clinical model (p = 0.042). The nomogram predicted T790M mutation with cutoff value was 0.69 and the score was above 130. CONCLUSION The nomogram developed in this study is a non-invasive, convenient, and economical method for predicting T790M mutation of lung adenocarcinoma, which has a good prospect for clinical application.
Collapse
Affiliation(s)
- Xiumei Li
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China
| | - Jianwei Chen
- Department of Radiology, Fujian Provincial Cancer Hospital, Fuzhou, Fujian, 350014, China
| | - Chengxiu Zhang
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai200062, China
| | - Zewen Han
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China
| | - Xiuying Zheng
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China
| | - Dairong Cao
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China.
- Department of Radiology, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350005, China.
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Shanghai200062, China.
| |
Collapse
|
27
|
Sun H, Zhou P, Chen G, Dai Z, Song P, Yao J. Radiomics nomogram for the prediction of Ki-67 index in advanced non-small cell lung cancer based on dual-phase enhanced computed tomography. J Cancer Res Clin Oncol 2023; 149:9301-9315. [PMID: 37204513 DOI: 10.1007/s00432-023-04856-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/13/2023] [Indexed: 05/20/2023]
Abstract
PURPOSE To develop a radiomics nomogram based on dual-phase enhanced computed tomography (CT) for predicting the Ki-67 index status in patients with advanced non-small cell lung cancer (NSCLC). METHODS 137 patients with NSCLC who had undergone dual-phase enhanced CT scans and Ki-67 examination within 2 weeks were retrospectively enrolled between January 2020 and December 2022. Clinical and laboratory data were collected, and the patients were categorized based on low or high expression of Ki-67 index, with a cut-off value of 40%. The cohort was randomly divided into a training group (n = 95) and a testing group (n = 42) at a ratio of 7:3. The least absolute shrinkage and selection operator (LASSO) algorithm was employed to select the most valuable radiomics features from the dual-phase enhanced CT images. Subsequently, a nomogram that incorporated the radiomics score and clinical factors associated with Ki-67 index status was established through univariate and multivariate logistic regression analyses. The predictive performance of the nomogram was evaluated using the area under the curve (AUC). RESULTS The AUC values of the radiomics features of artery phase and vein phase CT in the testing group were 0.748 and 0.758, respectively. The AUC of the dual-phase enhanced CT was 0.785, and the AUC of the developed nomogram was 0.859, which was higher than those of the radiomics (AUC, 0.785) and clinical models (AUC, 0.736). CONCLUSIONS The radiomics nomogram based on dual-phase enhanced CT images provides a promising method for predicting the Ki-67 index status in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Haitao Sun
- Medical Imaging Center, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, China
| | - Peng Zhou
- Medical Imaging Center, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, China
| | - Guoyue Chen
- Medical Imaging Center, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, China
| | - Zhengjun Dai
- Scientific Research Department of Huiying Medical Technology Co., Ltd, 66 Xixiaokou Road, Haidian District, Beijing, 100192, China
| | - Peiji Song
- Medical Imaging Center, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, China
| | - Jian Yao
- Medical Imaging Center, Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, China.
| |
Collapse
|
28
|
Hu B, Xia W, Piao S, Xiong J, Tang Y, Yu H, Tao G, Sun L, Shen M, Wagh A, Jaykel TJ, Zhang D, Li Y, Zhu L. A CT-based radiomics integrated model for discriminating pulmonary cryptococcosis granuloma from lung adenocarcinoma-a diagnostic test. Transl Lung Cancer Res 2023; 12:1790-1801. [PMID: 37691867 PMCID: PMC10483083 DOI: 10.21037/tlcr-23-389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023]
Abstract
Background Chest computed tomography (CT) is a critical tool in the diagnosis of pulmonary cryptococcosis as approximately 30% of normal immunity individuals may not exhibit any significant symptoms or laboratory findings. Pulmonary cryptococcosis granuloma and lung adenocarcinoma can appear similar on noncontrast chest CT. This study evaluates the use of an integrated model that was developed based on radiomic features combined with demographic and radiological features to differentiate pulmonary cryptococcosis nodules from lung adenocarcinomas. Methods Preoperative chest CT images for 215 patients with solid pulmonary nodules with histopathologically confirmed lung adenocarcinoma and cryptococcosis infection were collected from two clinical centers (108 cases in the training set and 107 cases in the test set divided by the different hospitals). Radiomics models were constructed based on nodular lesion volume (LV), 5-mm extended lesion volume (ELV), and perilesion volume (PLV). A demoradiological model was constructed using logistic regression based on demographic information (age, sex) and 12 radiological features (location, number, shape and specific imaging signs). Both models were used to build an integrated model, the performance of which was assessed using the test set. A junior and a senior radiologist evaluated the nodules. Receiver operating characteristic (ROC) curve analysis was conducted, and areas under the curve (AUCs), sensitivity (SEN), and specificity (SPE) of the models were calculated and compared. Results Among the radiomics models, AUCs of the LV, ELV, and PLV were 0.558, 0.757, and 0.470, respectively. Age, lesion number, and lobular sign were identified as independent discriminative features providing an AUC of 0.77 in the demoradiological model (SEN 0.815, SPE 0.642). The integrated model achieved the highest AUC of 0.801 (SEN 0.759, SPE 0.755), which was significantly higher than that obtained by a junior radiologist (AUC =0.689, P=0.024) but showed no significant difference from that of the senior radiologist (AUC =0.784, P=0.388). Conclusions An integrated model with radiomics and demoradiological features improves discrimination of cryptococcosis granulomas from solid adenocarcinomas on noncontrast CT. This model may be an effective strategy for machine complementation to discrimination by radiologists, and whole-lung automated recognition methods might dominate in the future.
Collapse
Affiliation(s)
- Bin Hu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Wei Xia
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Sirong Piao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ji Xiong
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Tang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hong Yu
- Department of Radiology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guangyu Tao
- Department of Radiology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Linlin Sun
- Department of Radiology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Minhui Shen
- College of Medical Instrument, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Ajay Wagh
- Section of Pulmonary and Critical Care Medicine/Interventional Pulmonology, The University of Chicago, Chicago, IL, USA
| | | | - Ding Zhang
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuxin Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Li Zhu
- Department of Radiology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
29
|
Tonneau M, Phan K, Manem VSK, Low-Kam C, Dutil F, Kazandjian S, Vanderweyen D, Panasci J, Malo J, Coulombe F, Gagné A, Elkrief A, Belkaïd W, Di Jorio L, Orain M, Bouchard N, Muanza T, Rybicki FJ, Kafi K, Huntsman D, Joubert P, Chandelier F, Routy B. Generalization optimizing machine learning to improve CT scan radiomics and assess immune checkpoint inhibitors' response in non-small cell lung cancer: a multicenter cohort study. Front Oncol 2023; 13:1196414. [PMID: 37546399 PMCID: PMC10400292 DOI: 10.3389/fonc.2023.1196414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
Background Recent developments in artificial intelligence suggest that radiomics may represent a promising non-invasive biomarker to predict response to immune checkpoint inhibitors (ICIs). Nevertheless, validation of radiomics algorithms in independent cohorts remains a challenge due to variations in image acquisition and reconstruction. Using radiomics, we investigated the importance of scan normalization as part of a broader machine learning framework to enable model external generalizability to predict ICI response in non-small cell lung cancer (NSCLC) patients across different centers. Methods Radiomics features were extracted and compared from 642 advanced NSCLC patients on pre-ICI scans using established open-source PyRadiomics and a proprietary DeepRadiomics deep learning technology. The population was separated into two groups: a discovery cohort of 512 NSCLC patients from three academic centers and a validation cohort that included 130 NSCLC patients from a fourth center. We harmonized images to account for variations in reconstruction kernel, slice thicknesses, and device manufacturers. Multivariable models, evaluated using cross-validation, were used to estimate the predictive value of clinical variables, PD-L1 expression, and PyRadiomics or DeepRadiomics for progression-free survival at 6 months (PFS-6). Results The best prognostic factor for PFS-6, excluding radiomics features, was obtained with the combination of Clinical + PD-L1 expression (AUC = 0.66 in the discovery and 0.62 in the validation cohort). Without image harmonization, combining Clinical + PyRadiomics or DeepRadiomics delivered an AUC = 0.69 and 0.69, respectively, in the discovery cohort, but dropped to 0.57 and 0.52, in the validation cohort. This lack of generalizability was consistent with observations in principal component analysis clustered by CT scan parameters. Subsequently, image harmonization eliminated these clusters. The combination of Clinical + DeepRadiomics reached an AUC = 0.67 and 0.63 in the discovery and validation cohort, respectively. Conversely, the combination of Clinical + PyRadiomics failed generalizability validations, with AUC = 0.66 and 0.59. Conclusion We demonstrated that a risk prediction model combining Clinical + DeepRadiomics was generalizable following CT scan harmonization and machine learning generalization methods. These results had similar performances to routine oncology practice using Clinical + PD-L1. This study supports the strong potential of radiomics as a future non-invasive strategy to predict ICI response in advanced NSCLC.
Collapse
Affiliation(s)
- Marion Tonneau
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
- Université de Médecine, Lille, France
| | - Kim Phan
- Imagia Canexia Health, Montreal, QC, Canada
| | - Venkata S. K. Manem
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
- Department of Mathematics and Computer Science, University of Quebec at Trois-Rivières, Trois-Rivières, QC, Canada
| | | | | | - Suzanne Kazandjian
- Department of Medical Oncology, Jewish General Hospital, Montreal, QC, Canada
| | - Davy Vanderweyen
- Department of Radiology, Centre Hospitalier de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - Justin Panasci
- Department of Medical Oncology, Jewish General Hospital, Montreal, QC, Canada
| | - Julie Malo
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
| | - François Coulombe
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
| | - Andréanne Gagné
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
| | - Arielle Elkrief
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
- Hemato-Oncology Division, Centre Hospitalier de l’université de Montreal, Montreal, QC, Canada
| | - Wiam Belkaïd
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
| | | | - Michele Orain
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
| | - Nicole Bouchard
- Department of Oncology, Centre Hospitalier de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - Thierry Muanza
- Department of Medical Oncology, Jewish General Hospital, Montreal, QC, Canada
- Department of Radiation Oncology, Lady Davis Institute of the Jewish General Hospital, Montreal, QC, Canada
| | | | - Kam Kafi
- Imagia Canexia Health, Montreal, QC, Canada
| | | | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec, Université Laval, Québec City, QC, Canada
- Department of Pathology, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
| | | | - Bertrand Routy
- Department of Cancer Research, Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM), Montreal, QC, Canada
- Hemato-Oncology Division, Centre Hospitalier de l’université de Montreal, Montreal, QC, Canada
| |
Collapse
|
30
|
Loi S, Mori M, Palumbo D, Crippa S, Palazzo G, Spezi E, Del Vecchio A, Falconi M, De Cobelli F, Fiorino C. Limited impact of discretization/interpolation parameters on the predictive power of CT radiomic features in a surgical cohort of pancreatic cancer patients. LA RADIOLOGIA MEDICA 2023:10.1007/s11547-023-01649-y. [PMID: 37289267 DOI: 10.1007/s11547-023-01649-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/15/2023] [Indexed: 06/09/2023]
Abstract
PURPOSE To explore the variation of the discriminative power of CT (Computed Tomography) radiomic features (RF) against image discretization/interpolation in predicting early distant relapses (EDR) after upfront surgery. MATERIALS AND METHODS Data of 144 patients with pre-surgical high contrast CT were processed consistently with IBSI (Image Biomarker Standardization Initiative) guidelines. Image interpolation/discretization parameters were intentionally changed, including cubic voxel size (0.21-27 mm3) and binning (32-128 grey levels) in a 15 parameter's sets. After excluding RF with poor inter-observer delineation agreement (ICC < 0.80) and not negligible inter-scanner variability, the variation of 80 RF against discretization/interpolation was first quantified. Then, their ability in classifying patients with early distant relapses (EDR, < 10 months, previously assessed at the first quartile value of time-to-relapse) was investigated in terms of AUC (Area Under Curve) variation for those RF significantly associated to EDR. RESULTS Despite RF variability against discretization/interpolation parameters was large and only 30/80 RF showed %COV < 20 (%COV = 100*STDEV/MEAN), AUC changes were relatively limited: for 30 RF significantly associated with EDR (AUC values around 0.60-0.70), the mean values of SD of AUC variability and AUC range were 0.02 and 0.05 respectively. AUC ranges were between 0.00 and 0.11, with values ≤ 0.05 in 16/30 RF. These variations were further reduced when excluding the extreme values of 32 and 128 for grey levels (Average AUC range 0.04, with values between 0.00 and 0.08). CONCLUSIONS The discriminative power of CT RF in the prediction of EDR after upfront surgery for pancreatic cancer is relatively invariant against image interpolation/discretization within a large range of voxel sizes and binning.
Collapse
Affiliation(s)
- Sara Loi
- Medical Physics, San Raffaele Scientific Institute, Via Olgettina 690, 20132, Milan, Italy
| | - Martina Mori
- Medical Physics, San Raffaele Scientific Institute, Via Olgettina 690, 20132, Milan, Italy
| | | | - Stefano Crippa
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute, Milan, Italy
| | - Gabriele Palazzo
- Medical Physics, San Raffaele Scientific Institute, Via Olgettina 690, 20132, Milan, Italy
| | - Emiliano Spezi
- School of Engineering, Cardiff University, Cardiff, Wales, UK
| | - Antonella Del Vecchio
- Medical Physics, San Raffaele Scientific Institute, Via Olgettina 690, 20132, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute, Milan, Italy
| | - Francesco De Cobelli
- San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute, Milan, Italy
| | - Claudio Fiorino
- Medical Physics, San Raffaele Scientific Institute, Via Olgettina 690, 20132, Milan, Italy.
| |
Collapse
|
31
|
Zhang R, Shi J, Liu S, Chen B, Li W. Performance of radiomics models derived from different CT reconstruction parameters for lung cancer risk prediction. BMC Pulm Med 2023; 23:132. [PMID: 37081469 PMCID: PMC10116652 DOI: 10.1186/s12890-023-02366-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 02/21/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND This study analysed the performance of radiomics features extracted from computed tomography (CT) images with different reconstruction parameters in differentiating malignant and benign pulmonary nodules. METHODS We evaluated routine chest CT images acquired from 148 participants with pulmonary nodules, which were pathologically diagnosed during surgery in West China Hospital, including a 5 mm unenhanced lung window, a 5 mm unenhanced mediastinal window, a 5 mm contrast-enhanced mediastinal window and a 1 mm unenhanced lung window. The pulmonary nodules were segmented, and 1409 radiomics features were extracted for each window. Then, we created 15 cohorts consisting of single windows or multiple windows. Univariate correlation analysis and principal component analysis were performed to select the features, and logistic regression analysis was performed to establish models for each cohort. The area under the curve (AUC) was applied to compare model performance. RESULTS There were 75 benign and 73 malignant pulmonary nodules, with mean diameters of 18.63 and 19.86 mm, respectively. For the single-window setting, the AUCs of the radiomics model from the 5 mm unenhanced lung window, 5 mm unenhanced mediastinal window, 5 mm contrast-enhanced mediastinal window and 1 mm unenhanced lung window were 0.771, 0.808, 0.750, and 0.771 in the training set and 0.711, 0.709, 0.684, and 0.674 in the test set, respectively. Regarding the multiple-window setting, the radiomics model based on all four windows showed an AUC of 0.825 in the training set and 0.743 in the test set. Statistically, the 15 models demonstrated comparable performances (P > 0.05). CONCLUSION A single chest CT window was acceptable in predicting the malignancy of pulmonary nodules, and additional windows did not statistically improve the performance of the radiomics models. In addition, slice thickness and contrast enhancement did not affect the diagnostic performance.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, 37 GuoXue Alley, Wuhou District, Chengdu, Sichuan Province, 610041, People's Republic of China
- Department of General Practice, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jie Shi
- GE Healthcare, Shanghai, China
| | | | - Bojiang Chen
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, 37 GuoXue Alley, Wuhou District, Chengdu, Sichuan Province, 610041, People's Republic of China.
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, 37 GuoXue Alley, Wuhou District, Chengdu, Sichuan Province, 610041, People's Republic of China.
| |
Collapse
|
32
|
Liu Y, Wei X, Feng X, Liu Y, Feng G, Du Y. Repeatability of radiomics studies in colorectal cancer: a systematic review. BMC Gastroenterol 2023; 23:125. [PMID: 37059990 PMCID: PMC10105401 DOI: 10.1186/s12876-023-02743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 03/22/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Recently, radiomics has been widely used in colorectal cancer, but many variable factors affect the repeatability of radiomics research. This review aims to analyze the repeatability of radiomics studies in colorectal cancer and to evaluate the current status of radiomics in the field of colorectal cancer. METHODS The included studies in this review by searching from the PubMed and Embase databases. Then each study in our review was evaluated using the Radiomics Quality Score (RQS). We analyzed the factors that may affect the repeatability in the radiomics workflow and discussed the repeatability of the included studies. RESULTS A total of 188 studies was included in this review, of which only two (2/188, 1.06%) studies controlled the influence of individual factors. In addition, the median score of RQS was 11 (out of 36), range-1 to 27. CONCLUSIONS The RQS score was moderately low, and most studies did not consider the repeatability of radiomics features, especially in terms of Intra-individual, scanners, and scanning parameters. To improve the generalization of the radiomics model, it is necessary to further control the variable factors of repeatability.
Collapse
Affiliation(s)
- Ying Liu
- School of Medical Imaging, North Sichuan Medical College, Sichuan Province, Nanchong City, 637000, China
| | - Xiaoqin Wei
- School of Medical Imaging, North Sichuan Medical College, Sichuan Province, Nanchong City, 637000, China
| | | | - Yan Liu
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China
| | - Guiling Feng
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China
| | - Yong Du
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China.
| |
Collapse
|
33
|
Deep learning-based harmonization of CT reconstruction kernels towards improved clinical task performance. Eur Radiol 2023; 33:2426-2438. [PMID: 36355196 DOI: 10.1007/s00330-022-09229-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/11/2022] [Accepted: 10/09/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVES To develop a deep learning-based harmonization framework, assessing whether it can improve performance of radiomics models given different kernels in different clinical tasks and additionally generalize to mitigate the effects of new/unobserved kernels on radiomics features. METHODS Patient data with 2 reconstruction kernels and phantom data with 22 reconstruction kernels were included. Eighty-five patients were studied for lymph node metastasis (LNM) prediction, and 164 patients for differential diagnosis between lung cancer (LC) and pulmonary tuberculosis (TB). Two convolutional neural network (CNN) models were developed to convert images (i) from B70f to B30f (CNNa) and (ii) from B30f to B70f (CNNb). Model performance between the two kernels was evaluated using AUC and compared with other well-known harmonization methods. Patient-normalized feature difference (PNFD) was used to identify the incompatible kernels (i.e., kernel with median PNFD > 1) with baseline (B30f/B70f), and measure the ability of the CNN models to convert the non-comparable kernels. RESULTS For LC versus pulmonary TB diagnosis, AUCs of CNNa vs. others were 0.85 vs. 0.54-0.74 (p = 0.0001-0.0003), and for CNNb vs. others: 0.87 vs. 0.54-0.86 (p = 0.0001-0.55). For LNM prediction, AUCs of CNNa vs. others were 0.68 vs. 0.56-0.61 (p = 0.10-0.39), and for CNNb vs. others: 0.78 vs. 0.70-0.73 (p = 0.07-0.40). After CNN harmonization, 17 of 20 (85%) of investigated unknown kernels produced comparable radiomics feature values relative to baseline (median PNFD from 1.10-2.31 to 0.23-1.13). CONCLUSION The CNN harmonization effectively improved performance of radiomics models between reconstruction kernels in different clinical tasks, and reduced feature differences between unknown kernels vs. baseline. KEY POINTS • The soft (B30f) and sharp (B70f) kernels strongly affect radiomics reproducibility and generalizability. • The convolutional neural network (CNN) harmonization methods performed better than location-scale (ComBat and centering-scaling) and matrix factorization harmonization methods (based on singular value decomposition (SVD) and independent component analysis (ICA)) in both clinical tasks. • The CNN harmonization methods improve feature reproducibility not only between specific kernels (B30f and B70f) from the same scanner, but also between unobserved kernels from different scanners of different vendors.
Collapse
|
34
|
Kiser K, Zhang J, Kim SG. Textural Features of Mouse Glioma Models Measured by Dynamic Contrast-Enhanced MR Images with 3D Isotropic Resolution. Tomography 2023; 9:721-735. [PMID: 37104129 PMCID: PMC10141208 DOI: 10.3390/tomography9020058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 04/28/2023] Open
Abstract
This paper investigates the effect of anisotropic resolution on the image textural features of pharmacokinetic (PK) parameters of a murine glioma model using dynamic contrast-enhanced (DCE) MR images acquired with an isotropic resolution at 7T with pre-contrast T1 mapping. The PK parameter maps of whole tumors at isotropic resolution were generated using the two-compartment exchange model combined with the three-site-two-exchange model. The textural features of these isotropic images were compared with those of simulated, thick-slice, anisotropic images to assess the influence of anisotropic voxel resolution on the textural features of tumors. The isotropic images and parameter maps captured distributions of high pixel intensity that were absent in the corresponding anisotropic images with thick slices. A significant difference was observed in 33% of the histogram and textural features extracted from anisotropic images and parameter maps, compared to those extracted from corresponding isotropic images. Anisotropic images in different orthogonal orientations demonstrated 42.1% of the histogram and textural features to be significantly different from those of isotropic images. This study demonstrates that the anisotropy of voxel resolution needs to be carefully considered when comparing the textual features of tumor PK parameters and contrast-enhanced images.
Collapse
Affiliation(s)
- Karl Kiser
- Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | |
Collapse
|
35
|
Khademi S, Heidarian S, Afshar P, Enshaei N, Naderkhani F, Rafiee MJ, Oikonomou A, Shafiee A, Babaki Fard F, plataniotis KN, Mohammadi A. Robust framework for COVID-19 identication from a multicenter dataset of chest CT scans. PLoS One 2023; 18:e0282121. [PMID: 36862633 PMCID: PMC9980818 DOI: 10.1371/journal.pone.0282121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 02/07/2023] [Indexed: 03/03/2023] Open
Abstract
The main objective of this study is to develop a robust deep learning-based framework to distinguish COVID-19, Community-Acquired Pneumonia (CAP), and Normal cases based on volumetric chest CT scans, which are acquired in different imaging centers using different scanners and technical settings. We demonstrated that while our proposed model is trained on a relatively small dataset acquired from only one imaging center using a specific scanning protocol, it performs well on heterogeneous test sets obtained by multiple scanners using different technical parameters. We also showed that the model can be updated via an unsupervised approach to cope with the data shift between the train and test sets and enhance the robustness of the model upon receiving a new external dataset from a different center. More specifically, we extracted the subset of the test images for which the model generated a confident prediction and used the extracted subset along with the training set to retrain and update the benchmark model (the model trained on the initial train set). Finally, we adopted an ensemble architecture to aggregate the predictions from multiple versions of the model. For initial training and development purposes, an in-house dataset of 171 COVID-19, 60 CAP, and 76 Normal cases was used, which contained volumetric CT scans acquired from one imaging center using a single scanning protocol and standard radiation dose. To evaluate the model, we collected four different test sets retrospectively to investigate the effects of the shifts in the data characteristics on the model's performance. Among the test cases, there were CT scans with similar characteristics as the train set as well as noisy low-dose and ultra-low-dose CT scans. In addition, some test CT scans were obtained from patients with a history of cardiovascular diseases or surgeries. This dataset is referred to as the "SPGC-COVID" dataset. The entire test dataset used in this study contains 51 COVID-19, 28 CAP, and 51 Normal cases. Experimental results indicate that our proposed framework performs well on all test sets achieving total accuracy of 96.15% (95%CI: [91.25-98.74]), COVID-19 sensitivity of 96.08% (95%CI: [86.54-99.5]), CAP sensitivity of 92.86% (95%CI: [76.50-99.19]), Normal sensitivity of 98.04% (95%CI: [89.55-99.95]) while the confidence intervals are obtained using the significance level of 0.05. The obtained AUC values (One class vs Others) are 0.993 (95%CI: [0.977-1]), 0.989 (95%CI: [0.962-1]), and 0.990 (95%CI: [0.971-1]) for COVID-19, CAP, and Normal classes, respectively. The experimental results also demonstrate the capability of the proposed unsupervised enhancement approach in improving the performance and robustness of the model when being evaluated on varied external test sets.
Collapse
Affiliation(s)
- Sadaf Khademi
- Concordia Institute for Information Systems Engineering, Concordia University, Montreal, Canada
| | - Shahin Heidarian
- Department of Electrical and Computer Engineering, Concordia University, Montreal, QC, Canada
| | - Parnian Afshar
- Concordia Institute for Information Systems Engineering, Concordia University, Montreal, Canada
| | - Nastaran Enshaei
- Concordia Institute for Information Systems Engineering, Concordia University, Montreal, Canada
| | - Farnoosh Naderkhani
- Concordia Institute for Information Systems Engineering, Concordia University, Montreal, Canada
| | - Moezedin Javad Rafiee
- Department of Medicine and Diagnostic Radiology, McGill University, Montreal, QC, Canada
| | - Anastasia Oikonomou
- Department of Medical Imaging, Sunnybrook Health Sciences Center, Toronto, Canada
| | - Akbar Shafiee
- Department of Cardiovascular Research, Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Arash Mohammadi
- Concordia Institute for Information Systems Engineering, Concordia University, Montreal, Canada
- * E-mail:
| |
Collapse
|
36
|
Liufu Y, Wen Y, Wu W, Su R, Liu S, Li J, Pan X, Chen K, Guan Y. Radiomics Analysis of Multiphasic Computed Tomography Images for Distinguishing High-Risk Thymic Epithelial Tumors From Low-Risk Thymic Epithelial Tumors. J Comput Assist Tomogr 2023; 47:220-228. [PMID: 36877755 DOI: 10.1097/rct.0000000000001407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
OBJECTIVES The objective of this study is to preoperatively investigate the value of multiphasic contrast-enhanced computed tomography (CT)-based radiomics signatures for distinguishing high-risk thymic epithelial tumors (HTET) from low-risk thymic epithelial tumors (LTET) compared with conventional CT signatures. MATERIALS AND METHODS Pathologically confirmed 305 thymic epithelial tumors (TETs), including 147 LTET (Type A/AB/B1) and 158 HTET (Type B2/B3/C), were retrospectively analyzed, and were randomly divided into training (n = 214) and validation cohorts (n = 91). All patients underwent nonenhanced, arterial contrast-enhanced, and venous contrast-enhanced CT analysis. The least absolute shrinkage and selection operator regression with 10-fold cross-validation was performed for radiomic models building, and multivariate logistic regression analysis was performed for radiological and combined models building. The performance of the model was evaluated by the area under the receiver operating characteristic curve (AUC of ROC), and the AUCs were compared using the Delong test. Decision curve analysis was used to evaluate the clinical value of each model. Nomogram and calibration curves were plotted for the combined model. RESULTS The AUCs for radiological model in the training and validation cohorts were 0.756 and 0.733, respectively. For nonenhanced, arterial contrast-enhanced, venous contrast-enhanced CT and 3-phase images combined radiomics models, the AUCs were 0.940, 0.946, 0.960, and 0.986, respectively, in the training cohort, whereas 0.859, 0.876, 0.930, and 0.923, respectively, in the validation cohort. The combined model, including CT morphology and radiomics signature, showed AUCs of 0.990 and 0.943 in the training and validation cohorts, respectively. Delong test and decision curve analysis showed that the predictive performance and clinical value of the 4 radiomics models and combined model were greater than the radiological model ( P < 0.05). CONCLUSIONS The combined model, including CT morphology and radiomics signature, greatly improved the predictive performance for distinguishing HTET from LTET. Radiomics texture analysis can be used as a noninvasive method for preoperative prediction of the pathological subtypes of TET.
Collapse
Affiliation(s)
- Yuling Liufu
- From the Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University
| | - Yanhua Wen
- From the Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University
| | - Wensheng Wu
- From the Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University
| | - Ruihua Su
- From the Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University
| | - Shuya Liu
- From the Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University
| | - Jingxu Li
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University
| | - Xiaohuan Pan
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University
| | - Kai Chen
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yubao Guan
- From the Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University
| |
Collapse
|
37
|
Yang M, Shi L, Huang T, Li G, Shao H, Shen Y, Zhu J, Ni B. Value of contrast-enhanced magnetic resonance imaging-T2WI-based radiomic features in distinguishing lung adenocarcinoma from lung squamous cell carcinoma with solid components >8 mm. J Thorac Dis 2023; 15:635-648. [PMID: 36910079 PMCID: PMC9992614 DOI: 10.21037/jtd-23-142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023]
Abstract
Background Radiomics is one of the research frontiers in the field of imaging and has excellent diagnostic performance. However, there is a lack of magnetic resonance imaging (MRI)-based omics studies on identifying pathological subtypes of lung cancer. Here we explored the value of the contrast-enhanced MRI-T2-weighted imaging (T2WI)-based radiomic analysis in distinguishing adenocarcinoma (Ade) from squamous cell carcinoma (Squ) with solid components >8 mm. Methods A retrospective analysis was performed of a total of 71 lung cancer patients who undergoing contrast-enhanced MRI and computed tomography (CT) before treatment, and the nodules had solid components ≥8 mm in our center from January 2020 to September 2021. All enrolled patients were divided into Squ and Ade groups according to the pathological results. In addition, the two groups were randomly divided into training set and validation set in a ratio of about 7:3. Radiomics software was used to extract the relevant radiomic features. The least absolute shrinkage and selection operator (Lasso) was used to screen radiomic features that were most relevant to lung cancer subtypes, thus calculating the radiomic scores (Rad-score) and constructing the radiomic models. Multivariate logistic regression was used to combine relevant clinical features with Rad-score to form combined model nomograms. The receiver operating characteristic (ROC) curves. the area under the ROC curve (AUC), the decision curve analysis (DCA) and the DeLong's test were used to evaluate the clinical application potentials. Results The sensitivity and specificity of the clinical model based on smoking was 75.0% and 93.8%. The AUC of the constructed magnetic resonance (MR)-Rad model for differentiating the pathological subtypes of lung cancer was 0.8651 in the validation sets. The AUC of the CT-Rad model in the validation set were 0.9286. The combined model constructed by combining clinical features and Rad-score had AUC of 0.8016, for identifying the 2 pathological subtypes of lung cancer in the validation set. There was no significant difference in diagnostic performance between MR-Rad model and CT-Rad model (P>0.05). Conclusions The MR-Rad model has a diagnostic performance similar to that of CT-Rad model, while the diagnostic performance of the combined mode was better than the single MR model.
Collapse
Affiliation(s)
- Maoyuan Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Shi
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tianwei Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guangzheng Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hancheng Shao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yijun Shen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bin Ni
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
38
|
Bologna M, Tenconi C, Corino VDA, Annunziata G, Orlandi E, Calareso G, Pignoli E, Valdagni R, Mainardi LT, Rancati T. Repeatability and reproducibility of MRI-radiomic features: A phantom experiment on a 1.5 T scanner. Med Phys 2023; 50:750-762. [PMID: 36310346 DOI: 10.1002/mp.16054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 11/11/2022] Open
Abstract
PURPOSE Aim of this study is to assess the repeatability of radiomic features on magnetic resonance images (MRI) and their stability to variations in time of repetition (TR), time of echo (TE), slice thickness (ST), and pixel spacing (PS) using vegetable phantoms. METHODS The organic phantom was realized using two cucumbers placed inside a cylindrical container, and the analysis was performed using T1-weighted (T1w), T2-weighted (T2w), and diffusion-weighted images. One dataset was used to test the repeatability of the radiomic features, whereas other four datasets were used to test the sensitivity of the different MRI sequences to image acquisition parameters (TR, TE, ST, and PS). Four regions of interest (ROIs) were segmented: two for the central part of each cucumber and two for the external parts. Radiomic features were extracted from each ROI using Pyradiomics. To assess the effect of preprocessing on the reduction of variability, features were extracted both before and after the preprocessing. The coefficient of variation (CV) and intra-class correlation coefficient (ICC) were used to evaluate variability. RESULTS The use of intensity standardization increased the stability for the first-order statistics features. Shape and size features were always stable for all the analyses. Textural features were particularly sensitive to changes in ST and PS, although some increase in stability could be obtained by voxel size resampling. When images underwent image preprocessing, the number of stable features (ICC > 0.75 and mean absolute CV < 0.3) was 33 for apparent diffusion coefficient (ADC), 52 for T1w, and 73 for T2w. CONCLUSIONS The most critical source of variability is related to changes in voxel size (either caused by changes in ST or PS). Preprocessing increases features stability to both test-retest and variation of the image acquisition parameters for all the types of analyzed MRI (T1w, T2w, and ADC), except for ST.
Collapse
Affiliation(s)
- Marco Bologna
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Chiara Tenconi
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy.,Department of Medical Physics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valentina D A Corino
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Gaetano Annunziata
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ester Orlandi
- Radiation Oncology 2, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppina Calareso
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Emanuele Pignoli
- Department of Medical Physics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Riccardo Valdagni
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy.,Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.,Department of Radiation Oncology 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luca T Mainardi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Tiziana Rancati
- Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
39
|
Li Y, Liu J, Yang X, Xu F, Wang L, He C, Lin L, Qing H, Ren J, Zhou P. Radiomic and quantitative-semantic models of low-dose computed tomography for predicting the poorly differentiated invasive non-mucinous pulmonary adenocarcinoma. LA RADIOLOGIA MEDICA 2023; 128:191-202. [PMID: 36637740 DOI: 10.1007/s11547-023-01591-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/04/2023] [Indexed: 01/14/2023]
Abstract
PURPOSE Poorly differentiated invasive non-mucinous pulmonary adenocarcinoma (IPA), based on the novel grading system, was related to poor prognosis, with a high risk of lymph node metastasis and local recurrence. This study aimed to build the radiomic and quantitative-semantic models of low-dose computed tomography (LDCT) to preoperatively predict the poorly differentiated IPA in nodules with solid component, and compare their diagnostic performance with radiologists. MATERIALS AND METHODS A total of 396 nodules from 388 eligible patients, who underwent LDCT scan within 2 weeks before surgery and were pathologically diagnosed with IPA, were retrospectively enrolled between July 2018 and December 2021. Nodules were divided into two independent cohorts according to scanners: primary cohort (195 well/moderate differentiated and 64 poorly differentiated) and validation cohort (104 well/moderate differentiated and 33 poorly differentiated). The radiomic and quantitative-semantic models were built using multivariable logistic regression. The diagnostic performance of the models and radiologists was assessed by area under curve (AUC) of receiver operating characteristic (ROC) curve, accuracy, sensitivity, and specificity. RESULTS No significant differences of AUCs were found between the radiomic and quantitative-semantic model in primary and validation cohorts (0.921 vs. 0.923, P = 0.846 and 0.938 vs. 0.911, P = 0.161). Both the models outperformed three radiologists in the validation cohort (all P < 0.05). CONCLUSIONS The radiomic and quantitative-semantic models of LDCT, which could identify the poorly differentiated IPA with excellent diagnostic performance, might provide guidance for therapeutic decision making, such as choosing appropriate surgical method or adjuvant chemotherapy.
Collapse
Affiliation(s)
- Yong Li
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No.55, Section 4, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Jieke Liu
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No.55, Section 4, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Xi Yang
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No.55, Section 4, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Fuyang Xu
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No.55, Section 4, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Lu Wang
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No.55, Section 4, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Changjiu He
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No.55, Section 4, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Libo Lin
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No.55, Section 4, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Haomiao Qing
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No.55, Section 4, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Jing Ren
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No.55, Section 4, South Renmin Road, Chengdu, 610041, Sichuan, China
| | - Peng Zhou
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No.55, Section 4, South Renmin Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
40
|
Yuan H, Kang B, Sun K, Qin S, Ji C, Wang X. CT-based radiomics nomogram for differentiation of adrenal hyperplasia from lipid-poor adenoma: an exploratory study. BMC Med Imaging 2023; 23:4. [PMID: 36611159 PMCID: PMC9826591 DOI: 10.1186/s12880-022-00951-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/14/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND To establish and verify a radiomics nomogram for differentiating isolated micronodular adrenal hyperplasia (iMAD) from lipid-poor adenoma (LPA) based on computed tomography (CT)-extracted radiomic features. METHODS A total of 148 patients with iMAD or LPA were divided into three cohorts: a training cohort (n = 72; 37 iMAD and 35 LPA), a validation cohort (n = 36; 22 iMAD and 14 LPA), and an external validation cohort (n = 40; 20 iMAD and 20 LPA). Radiomics features were extracted from contrast-enhanced and non-contrast CT images. The least absolute shrinkage and selection operator (LASSO) method was applied to develop a triphasic radiomics model and unenhanced radiomics model using reproducible radiomics features. A clinical model was constructed using certain laboratory variables and CT findings. Radiomics nomogram was established by selected radiomics signature and clinical factors. Nomogram performance was assessed by calibration curve, the areas under receiver operating characteristic curves (AUC), and decision curve analysis (DCA). RESULTS Eleven and eight extracted features were finally selected to construct an unenhanced radiomics model and a triphasic radiomics model, respectively. There was no significant difference in AUC between the two models in the external validation cohort (0.838 vs. 0.843, p = 0.949). The radiomics nomogram inclusive of the unenhanced model, maximum diameter, and aldosterone showed the AUC of 0.951, 0.938, and 0.893 for the training, validation, and external validation cohorts, respectively. The nomogram showed good calibration, and the DCA demonstrated the superiority of the nomogram compared with the clinical factors model alone in terms of clinical usefulness. CONCLUSIONS A radiomics nomogram based on unenhanced CT images and clinical variables showed favorable performance for distinguishing iMAD from LPA. In addition, an efficient unenhanced model can help avoid extra contrast-enhanced scanning and radiation risk.
Collapse
Affiliation(s)
- Hongtao Yuan
- grid.27255.370000 0004 1761 1174Shandong Provincial Hospital, Shandong University, Jinan, Shandong China
| | - Bing Kang
- grid.460018.b0000 0004 1769 9639Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong China
| | - Kui Sun
- grid.460018.b0000 0004 1769 9639Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong China ,grid.410638.80000 0000 8910 6733School of Medicine, Shandong First Medical University, Jinan, Shandong China
| | - Songnan Qin
- grid.27255.370000 0004 1761 1174Shandong Provincial Hospital, Shandong University, Jinan, Shandong China
| | - Congshan Ji
- grid.460018.b0000 0004 1769 9639Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong China
| | - Ximing Wang
- grid.460018.b0000 0004 1769 9639Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong China
| |
Collapse
|
41
|
Zhang X, Lu B, Yang X, Lan D, Lin S, Zhou Z, Li K, Deng D, Peng P, Zeng Z, Long L. Prognostic analysis and risk stratification of lung adenocarcinoma undergoing EGFR-TKI therapy with time-serial CT-based radiomics signature. Eur Radiol 2023; 33:825-835. [PMID: 36166088 PMCID: PMC9889474 DOI: 10.1007/s00330-022-09123-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/05/2022] [Accepted: 08/19/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVES To evaluate the value of time-serial CT radiomics features in predicting progression-free survival (PFS) for lung adenocarcinoma (LUAD) patients after epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) therapy. MATERIALS AND METHODS LUAD patients treated with EGFR-TKIs were retrospectively included from three independent institutes and divided into training and validation cohorts. Intratumoral and peritumoral features were extracted from time-serial non-contrast chest CT (including pre-therapy and first follow-up images); moreover, the percentage variation per unit time (day) was introduced to adjust for the different follow-up periods of each patient. Test-retest was performed to exclude irreproducible features, while the Boruta algorithm was used to select critical radiomics features. Radiomics signatures were constructed with random forest survival models in the training cohort and compared against baseline clinical characteristics through Cox regression and nonparametric testing of concordance indices (C-indices). RESULTS The training cohort included 131 patients (74 women, 56.5%) from one institute and the validation cohort encompassed 41 patients (24 women, 58.5%) from two other institutes. The optimal signature contained 10 features and 7 were unit time feature variations. The comprehensive radiomics model outperformed the pre-therapy clinical characteristics in predicting PFS (training: 0.78, 95% CI: [0.72, 0.84] versus 0.55, 95% CI: [0.49, 0.62], p < 0.001; validation: 0.72, 95% CI: [0.60, 0.84] versus 0.54, 95% CI: [0.42, 0.66], p < 0.001). CONCLUSION Radiomics signature derived from time-serial CT images demonstrated optimal prognostic performance of disease progression. This dynamic imaging biomarker holds the promise of monitoring treatment response and achieving personalized management. KEY POINTS • The intrinsic tumor heterogeneity can be highly dynamic under the therapeutic effect of EGFR-TKI treatment, and the inevitable development of drug resistance may disrupt the duration of clinical benefit. Decision-making remained challenging in practice to detect the emergence of acquired resistance during the early response phase. • Time-serial CT-based radiomics signature integrating intra- and peritumoral features offered the potential to predict progression-free survival for LUAD patients treated with EGFR-TKIs. • The dynamic imaging signature allowed for prognostic risk stratification.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| | - Bingfeng Lu
- Department of Radiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi China
| | - Xinguan Yang
- Department of Radiology, Guilin People’s Hospital, Guilin, Guangxi China
| | - Dong Lan
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi China
| | | | - Zhipeng Zhou
- Department of Radiology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi China
| | - Kai Li
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| | - Dong Deng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| | - Peng Peng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| | - Zisan Zeng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| | - Liling Long
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu District, Nanning, 530021 Guangxi China
| |
Collapse
|
42
|
CT-Based Radiomics Can Predict the Efficacy of Anlotinib in Advanced Non-Small-Cell Lung Cancer. JOURNAL OF ONCOLOGY 2022; 2022:4182540. [PMID: 36600966 PMCID: PMC9807313 DOI: 10.1155/2022/4182540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/27/2022]
Abstract
Anlotinib is a small-molecule RTK inhibitor that has achieved certain results in further-line treatment, but many patients do not respond to this drug and lack effective methods for identification. Although radiomics has been widely used in lung cancer, very few studies have been conducted in the field of antiangiogenic drugs. This study aims to develop a new model to predict the efficacy of patients receiving anlotinib by combining pretreatment computed tomography (CT) radiomic characters with clinical characters, in order to assist precision medicine of pulmonary cancer. 254 patients from seven institutions were involved in the study. Lesions were selected according to the RECIST 1.1 criteria, and the corresponding radiomic features were obtained. We constructed prediction models based on clinical, NCE-CT, and CE-CT radiomic features, respectively, and evaluated the prediction performance of the models for training sets, internal validation sets, and external validation sets. In the RAD score only model, the area under curve(AUC) of the NCE-CT cohort was 0.740 (95% CI: 0.622, 0.857) for the training set, 0.711 (95% CI: 0.480, 0.942) for the internal validation set, and 0.633(95% CI: 0.479, 0.787) for the external validation set, while that of the CE-CT cohort was 0.815 (95% CI: 0.705, 0.926) for the training set, 0.771 (95% CI: 0.539, 1.000) for the internal validation set, and 0.701 (95% CI: 0.489, 0.913) for the external validation set. In the RAD score-combined model, the AUC of the NCE-CT cohort was 0.796 (95% CI: 0.691, 0.901) for the training set, 0.579 (95% CI: 0.309, 0.848) for the internal validation set, and 0.590 (95% CI: 0.427, 0.753) for the external validation set, while that of the CE-CT cohort was 0.902 (95% CI: 0.828, 0.977) for the training set, 0.865 (95% CI: 0.696, 1.000) for the internal validation set, and 0.837 (95% CI: 0.682, 0.992) for the external validation set. In conclusion, radiomics has accurate predictions for the efficacy of anlotinib. CE-CT-based radiomic models have the best predictive potential in predicting the efficacy of anlotinib, and model predictions become better when they are combined with clinical characteristics.
Collapse
|
43
|
Chen Z, Yi L, Peng Z, Zhou J, Zhang Z, Tao Y, Lin Z, He A, Jin M, Zuo M. Development and validation of a radiomic nomogram based on pretherapy dual-energy CT for distinguishing adenocarcinoma from squamous cell carcinoma of the lung. Front Oncol 2022; 12:949111. [PMID: 36505773 PMCID: PMC9727167 DOI: 10.3389/fonc.2022.949111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 10/26/2022] [Indexed: 11/24/2022] Open
Abstract
Objective Based on pretherapy dual-energy computed tomography (DECT) images, we developed and validated a nomogram combined with clinical parameters and radiomic features to predict the pathologic subtypes of non-small cell lung cancer (NSCLC) - adenocarcinoma (ADC) and squamous cell carcinoma (SCC). Methods A total of 129 pathologically confirmed NSCLC patients treated at the Second Affiliated Hospital of Nanchang University from October 2017 to October 2021 were retrospectively analyzed. Patients were randomly divided in a ratio of 7:3 (n=90) into training and validation cohorts (n=39). Patients' pretherapy clinical parameters were recorded. Radiomics features of the primary lesion were extracted from two sets of monoenergetic images (40 keV and 100 keV) in arterial phases (AP) and venous phases (VP). Features were selected successively through the intra-class correlation coefficient (ICC) and the least absolute shrinkage and selection operator (LASSO). Multivariate logistic regression analysis was then performed to establish predictive models. The prediction performance between models was evaluated and compared using the receiver operating characteristic (ROC) curve, DeLong test, and Akaike information criterion (AIC). A nomogram was developed based on the model with the best predictive performance to evaluate its calibration and clinical utility. Results A total of 87 ADC and 42 SCC patients were enrolled in this study. Among the five constructed models, the integrative model (AUC: Model 4 = 0.92, Model 5 = 0.93) combining clinical parameters and radiomic features had a higher AUC than the individual clinical models or radiomic models (AUC: Model 1 = 0.84, Model 2 = 0.79, Model 3 = 0.84). The combined clinical-venous phase radiomics model had the best predictive performance, goodness of fit, and parsimony; the area under the ROC curve (AUC) of the training and validation cohorts was 0.93 and 0.90, respectively, and the AIC value was 60.16. Then, this model was visualized as a nomogram. The calibration curves demonstrated it's good calibration, and decision curve analysis (DCA) proved its clinical utility. Conclusion The combined clinical-radiomics model based on pretherapy DECT showed good performance in distinguishing ADC and SCC of the lung. The nomogram constructed based on the best-performing combined clinical-venous phase radiomics model provides a relatively accurate, convenient and noninvasive method for predicting the pathological subtypes of ADC and SCC in NSCLC.
Collapse
Affiliation(s)
- Zhiyong Chen
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Li Yi
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhiwei Peng
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianzhong Zhou
- Department of Radiology, The Quzhou City People’s Hospital, Quzhou, Zhejiang, China
| | - Zhaotao Zhang
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yahong Tao
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ze Lin
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Anjing He
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mengni Jin
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Minjing Zuo
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China,*Correspondence: Minjing Zuo,
| |
Collapse
|
44
|
Sun Y, Zhao W, Kuang K, Jin L, Gao P, Duan S, Xiao Y, Liu J, Li M. Non-contrast and contrast enhanced computed tomography radiomics in preoperative discrimination of lung invasive and non-invasive adenocarcinoma. Front Med (Lausanne) 2022; 9:939434. [PMID: 36405608 PMCID: PMC9672504 DOI: 10.3389/fmed.2022.939434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 10/13/2022] [Indexed: 01/21/2025] Open
Abstract
OBJECTIVE This study aimed to assess the value of radiomics based on non-contrast computed tomography (NCCT) and contrast-enhanced computed tomography (CECT) images in the preoperative discrimination between lung invasive adenocarcinomas (IAC) and non-invasive adenocarcinomas (non-IAC). METHODS We enrolled 1,185 pulmonary nodules (478 non-IACs and 707 IACs) to build and validate radiomics models. An external testing set comprising 63 pulmonary nodules was collected to verify the generalization of the models. Radiomic features were extracted from both NCCT and CECT images. The predictive performance of radiomics models in the validation and external testing sets were evaluated and compared with radiologists' evaluations. The predictive performances of the radiomics models were also compared between three subgroups in the validation set (Group 1: solid nodules, Group 2: part-solid nodules, and Group 3: pure ground-glass nodules). RESULTS The NCCT, CECT, and combined models showed good ability to discriminate between IAC and non-IAC [respective areas under the curve (AUCs): validation set = 0.91, 0.90, and 0.91; Group 1 = 0.82, 0.79, and 0.81; Group 2 = 0.93, 0.92, and 0.93; and Group 3 = 0.90, 0.90, and 0.89]. In the external testing set, the AUC of the three models were 0.89, 0.91, and 0.89, respectively. The accuracies of these three models were comparable to those of the senior radiologist and better those that of the junior radiologist. CONCLUSION Radiomic models based on CT images showed good predictive performance in discriminating between lung IAC and non-IAC, especially in part solid nodule group. However, radiomics based on CECT images provided no additional value compared to NCCT images.
Collapse
Affiliation(s)
- Yingli Sun
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Wei Zhao
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, China
| | | | - Liang Jin
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Pan Gao
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | | | - Yi Xiao
- Department of Radiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming Li
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
45
|
Peng G, Zhan Y, Wu Y, Zeng C, Wang S, Guo L, Liu W, Luo L, Wang R, Huang K, Huang B, Chen J, Chen C. Radiomics models based on CT at different phases predicting lymph node metastasis of esophageal squamous cell carcinoma (GASTO-1089). Front Oncol 2022; 12:988859. [PMID: 36387160 PMCID: PMC9643555 DOI: 10.3389/fonc.2022.988859] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/07/2022] [Indexed: 02/05/2023] Open
Abstract
PURPOSE To investigate the value of radiomics models based on CT at different phases (non-contrast-enhanced and contrast-enhanced images) in predicting lymph node (LN) metastasis in esophageal squamous cell carcinoma (ESCC). METHODS AND MATERIALS Two hundred and seventy-four eligible patients with ESCC were divided into a training set (n =193) and a validation set (n =81). The least absolute shrinkage and selection operator algorithm (LASSO) was used to select radiomics features. The predictive models were constructed with radiomics features and clinical factors through multivariate logistic regression analysis. The predictive performance and clinical application value of the models were evaluated by area under receiver operating characteristic curve (AUC) and decision curve analysis (DCA). The Delong Test was used to evaluate the differences in AUC among models. RESULTS Sixteen and eighteen features were respectively selected from non-contrast-enhanced CT (NECT) and contrast-enhanced CT (CECT) images. The model established using only clinical factors (Model 1) has an AUC value of 0.655 (95%CI 0.552-0.759) with a sensitivity of 0.585, a specificity of 0.725 and an accuracy of 0.654. The models contained clinical factors with radiomics features of NECT or/and CECT (Model 2,3,4) have significantly improved prediction performance. The values of AUC of Model 2,3,4 were 0.766, 0.811 and 0.809, respectively. It also achieved a great AUC of 0.800 in the model built with only radiomics features derived from NECT and CECT (Model 5). DCA suggested the potential clinical benefit of model prediction of LN metastasis of ESCC. A comparison of the receiver operating characteristic (ROC) curves using the Delong test indicated that Models 2, 3, 4, and 5 were superior to Model 1(P< 0.05), and no difference was found among Model 2, 3, 4 and Model 5(P > 0.05). CONCLUSION Radiomics models based on CT at different phases could accurately predict the lymph node metastasis in patients with ESCC, and their predictive efficiency was better than the clinical model based on tumor size criteria. NECT-based radiomics model could be a reasonable option for ESCC patients due to its lower price and availability for renal failure or allergic patients.
Collapse
Affiliation(s)
- Guobo Peng
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Radiation Oncology, Meizhou People’s Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou, China
| | - Yizhou Zhan
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yanxuan Wu
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Chengbing Zeng
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Siyan Wang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Longjia Guo
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Weitong Liu
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Radiation Oncology, Meizhou People’s Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou, China
| | - Limei Luo
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Ruoheng Wang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Kang Huang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Baotian Huang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Jianzhou Chen
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Chuangzhen Chen
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
46
|
Zhao W, Xiong Z, Tian D, Wang K, Zhao M, Lu X, Qin D, Li Z. The adding value of contrast-enhanced CT radiomics: Differentiating tuberculosis from non-tuberculous infectious lesions presenting as solid pulmonary nodules or masses. Front Public Health 2022; 10:1018527. [PMID: 36267999 PMCID: PMC9577178 DOI: 10.3389/fpubh.2022.1018527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/20/2022] [Indexed: 01/28/2023] Open
Abstract
Purpose To compare the value of contrast-enhanced CT (CECT) and non-contrast-enhanced CT (NCECT) radiomics models in differentiating tuberculosis (TB) from non-tuberculous infectious lesions (NTIL) presenting as solid pulmonary nodules or masses, and develop a combine radiomics model (RM). Materials and methods This study was a retrospective analysis of 101 lesions in 95 patients, including 49 lesions (from 45 patients) in the TB group and 52 lesions (from 50 patients) in the NTIL group. Lesions were randomly divided into training and test sets in the ratio of 7:3. Conventional imaging features were used to construct a conventional imaging model (IM). Radiomics features screening and NCECT or CECT RM construction were carried out by correlation analysis and gradient boosting decision tree, and logistic regression. Finally, conventional IM, NCECT RM, and CECT RM were used for combine RM construction. Additionally, we recruited three radiologists for independent diagnosis. The differential diagnostic performance of each model was assessed using the areas under the receiver operating characteristic curve (AUCs). Results The CECT RM (training AUC, 0.874; test AUC, 0.796) outperformed the conventional IM (training AUC, 0.792; test AUC, 0.708), the NCECT RM (training AUC, 0.835; test AUC, 0.704), and three radiologists. The diagnostic efficacy of the combine RM (training AUC, 0.922; test AUC, 0.833) was best in the training and test sets. Conclusions The diagnostic efficacy of the CECT RM was superior to that of the NCECT RM in identifying TB from NTIL presenting as solid pulmonary nodules or masses. The combine RM had the best performance and may outperform expert radiologists.
Collapse
Affiliation(s)
- Wenjing Zhao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ziqi Xiong
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Di Tian
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Kunpeng Wang
- Department of Radiology, Dalian Public Health Clinical Center, Dalian, China
| | | | - Xiwei Lu
- Department of Tuberculosis, Dalian Public Health Clinical Center, Dalian, China
| | - Dongxue Qin
- Department of Radiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China,*Correspondence: Dongxue Qin
| | - Zhiyong Li
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China,Zhiyong Li
| |
Collapse
|
47
|
Ibrahim A, Lu L, Yang H, Akin O, Schwartz LH, Zhao B. The Impact of Image Acquisition Parameters and ComBat Harmonization on the Predictive Performance of Radiomics: A Renal Cell Carcinoma Model. APPLIED SCIENCES (BASEL, SWITZERLAND) 2022; 12:9824. [PMID: 37091743 PMCID: PMC10121203 DOI: 10.3390/app12199824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Radiomics, one of the potential methods for developing clinical biomarker, is one of the exponentially growing research fields. In addition to its potential, several limitations have been identified in this field, and most importantly the effects of variations in imaging parameters on radiomic features (RFs). In this study, we investigate the potential of RFs to predict overall survival in patients with clear cell renal cell carcinoma, as well as the impact of ComBat harmonization on the performance of RF models. We assessed the robustness of the results by performing the analyses a thousand times. Publicly available CT scans of 179 patients were retrospectively collected and analyzed. The scans were acquired using different imaging vendors and parameters in different medical centers. The performance was calculated by averaging the metrics over all runs. On average, the clinical model significantly outperformed the radiomic models. The use of ComBat harmonization, on average, did not significantly improve the performance of radiomic models. Hence, the variability in image acquisition and reconstruction parameters significantly affect the performance of radiomic models. The development of radiomic specific harmonization techniques remain a necessity for the advancement of the field.
Collapse
Affiliation(s)
- Abdalla Ibrahim
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Correspondence:
| | - Lin Lu
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hao Yang
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Oguz Akin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lawrence H. Schwartz
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Binsheng Zhao
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
48
|
Dercle L, McGale J, Sun S, Marabelle A, Yeh R, Deutsch E, Mokrane FZ, Farwell M, Ammari S, Schoder H, Zhao B, Schwartz LH. Artificial intelligence and radiomics: fundamentals, applications, and challenges in immunotherapy. J Immunother Cancer 2022; 10:jitc-2022-005292. [PMID: 36180071 PMCID: PMC9528623 DOI: 10.1136/jitc-2022-005292] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 11/04/2022] Open
Abstract
Immunotherapy offers the potential for durable clinical benefit but calls into question the association between tumor size and outcome that currently forms the basis for imaging-guided treatment. Artificial intelligence (AI) and radiomics allow for discovery of novel patterns in medical images that can increase radiology’s role in management of patients with cancer, although methodological issues in the literature limit its clinical application. Using keywords related to immunotherapy and radiomics, we performed a literature review of MEDLINE, CENTRAL, and Embase from database inception through February 2022. We removed all duplicates, non-English language reports, abstracts, reviews, editorials, perspectives, case reports, book chapters, and non-relevant studies. From the remaining articles, the following information was extracted: publication information, sample size, primary tumor site, imaging modality, primary and secondary study objectives, data collection strategy (retrospective vs prospective, single center vs multicenter), radiomic signature validation strategy, signature performance, and metrics for calculation of a Radiomics Quality Score (RQS). We identified 351 studies, of which 87 were unique reports relevant to our research question. The median (IQR) of cohort sizes was 101 (57–180). Primary stated goals for radiomics model development were prognostication (n=29, 33.3%), treatment response prediction (n=24, 27.6%), and characterization of tumor phenotype (n=14, 16.1%) or immune environment (n=13, 14.9%). Most studies were retrospective (n=75, 86.2%) and recruited patients from a single center (n=57, 65.5%). For studies with available information on model testing, most (n=54, 65.9%) used a validation set or better. Performance metrics were generally highest for radiomics signatures predicting treatment response or tumor phenotype, as opposed to immune environment and overall prognosis. Out of a possible maximum of 36 points, the median (IQR) of RQS was 12 (10–16). While a rapidly increasing number of promising results offer proof of concept that AI and radiomics could drive precision medicine approaches for a wide range of indications, standardizing the data collection as well as optimizing the methodological quality and rigor are necessary before these results can be translated into clinical practice.
Collapse
Affiliation(s)
- Laurent Dercle
- Radiology, NewYork-Presbyterian/Columbia University Medical Center, New York, New York, USA
| | - Jeremy McGale
- Radiology, NewYork-Presbyterian/Columbia University Medical Center, New York, New York, USA
| | - Shawn Sun
- Radiology, NewYork-Presbyterian/Columbia University Medical Center, New York, New York, USA
| | - Aurelien Marabelle
- Therapeutic Innovation and Early Trials, Gustave Roussy, Villejuif, Île-de-France, France
| | - Randy Yeh
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Eric Deutsch
- Radiation Oncology, Gustave Roussy, Villejuif, Île-de-France, France
| | | | - Michael Farwell
- Division of Nuclear Medicine and Molecular Imaging, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Samy Ammari
- Radiation Oncology, Gustave Roussy, Villejuif, Île-de-France, France.,Radiology, Institut de Cancérologie Paris Nord, Sarcelles, France
| | - Heiko Schoder
- Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Binsheng Zhao
- Radiology, NewYork-Presbyterian/Columbia University Medical Center, New York, New York, USA
| | - Lawrence H Schwartz
- Radiology, NewYork-Presbyterian/Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
49
|
Chen Q, Zhang L, Liu S, You J, Chen L, Jin Z, Zhang S, Zhang B. Radiomics in precision medicine for gastric cancer: opportunities and challenges. Eur Radiol 2022; 32:5852-5868. [PMID: 35316364 DOI: 10.1007/s00330-022-08704-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/20/2022] [Accepted: 02/28/2022] [Indexed: 01/19/2023]
Abstract
OBJECTIVES Radiomic features derived from routine medical images show great potential for personalized medicine in gastric cancer (GC). We aimed to evaluate the current status and quality of radiomic research as well as its potential for identifying biomarkers to predict therapy response and prognosis in patients with GC. METHODS We performed a systematic search of the PubMed and Embase databases for articles published from inception through July 10, 2021. The phase classification criteria for image mining studies and the radiomics quality scoring (RQS) tool were applied to evaluate scientific and reporting quality. RESULTS Twenty-five studies consisting of 10,432 patients were included. 96% of studies extracted radiomic features from CT images. Association between radiomic signature and therapy response was evaluated in seven (28%) studies; association with survival was evaluated in 17 (68%) studies; one (4%) study analyzed both. All results of the included studies showed significant associations. Based on the phase classification criteria for image mining studies, 18 (72%) studies were classified as phase II, with two, four, and one studies as discovery science, phase 0 and phase I, respectively. The median RQS score for the radiomic studies was 44.4% (range, 0 to 55.6%). There was extensive heterogeneity in the study population, tumor stage, treatment protocol, and radiomic workflow amongst the studies. CONCLUSIONS Although radiomic research in GC is highly heterogeneous and of relatively low quality, it holds promise for predicting therapy response and prognosis. Efforts towards standardization and collaboration are needed to utilize radiomics for clinical application. KEY POINTS • Radiomics application of gastric cancer is increasingly being reported, particularly in predicting therapy response and survival. • Although radiomics research in gastric cancer is highly heterogeneous and relatively low quality, it holds promise for predicting clinical outcomes. • Standardized imaging protocols and radiomic workflow are needed to facilitate radiomics into clinical use.
Collapse
Affiliation(s)
- Qiuying Chen
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Lu Zhang
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Shuyi Liu
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Jingjing You
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Luyan Chen
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Zhe Jin
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.,Graduate College, Jinan University, Guangzhou, Guangdong, China
| | - Shuixing Zhang
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China. .,Graduate College, Jinan University, Guangzhou, Guangdong, China.
| | - Bin Zhang
- Department of Radiology, the First Affiliated Hospital, Jinan University, No.613, Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China. .,Graduate College, Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
50
|
Keek SA, Kayan E, Chatterjee A, Belderbos JSA, Bootsma G, van den Borne B, Dingemans AMC, Gietema HA, Groen HJM, Herder J, Pitz C, Praag J, De Ruysscher D, Schoenmaekers J, Smit HJM, Stigt J, Westenend M, Zeng H, Woodruff HC, Lambin P, Hendriks L. Investigation of the added value of CT-based radiomics in predicting the development of brain metastases in patients with radically treated stage III NSCLC. Ther Adv Med Oncol 2022; 14:17588359221116605. [PMID: 36032350 PMCID: PMC9403451 DOI: 10.1177/17588359221116605] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/12/2022] [Indexed: 12/04/2022] Open
Abstract
Introduction: Despite radical intent therapy for patients with stage III non-small-cell
lung cancer (NSCLC), cumulative incidence of brain metastases (BM) reaches
30%. Current risk stratification methods fail to accurately identify these
patients. As radiomics features have been shown to have predictive value,
this study aims to develop a model combining clinical risk factors with
radiomics features for BM development in patients with radically treated
stage III NSCLC. Methods: Retrospective analysis of two prospective multicentre studies. Inclusion
criteria: adequately staged [18F-fluorodeoxyglucose positron
emission tomography-computed tomography (18-FDG-PET-CT), contrast-enhanced
chest CT, contrast-enhanced brain magnetic resonance imaging/CT] and
radically treated stage III NSCLC, exclusion criteria: second primary within
2 years of NSCLC diagnosis and prior prophylactic cranial irradiation.
Primary endpoint was BM development any time during follow-up (FU). CT-based
radiomics features (N = 530) were extracted from the
primary lung tumour on 18-FDG-PET-CT images, and a list of clinical features
(N = 8) was collected. Univariate feature selection
based on the area under the curve (AUC) of the receiver operating
characteristic was performed to identify relevant features. Generalized
linear models were trained using the selected features, and multivariate
predictive performance was assessed through the AUC. Results: In total, 219 patients were eligible for analysis. Median FU was 59.4 months
for the training cohort and 67.3 months for the validation cohort; 21 (15%)
and 17 (22%) patients developed BM in the training and validation cohort,
respectively. Two relevant clinical features (age and adenocarcinoma
histology) and four relevant radiomics features were identified as
predictive. The clinical model yielded the highest AUC value of 0.71 (95%
CI: 0.58–0.84), better than radiomics or a combination of clinical
parameters and radiomics (both an AUC of 0.62, 95% CIs of 0.47–076 and
0.48–0.76, respectively). Conclusion: CT-based radiomics features of primary NSCLC in the current setup could not
improve on a model based on clinical predictors (age and adenocarcinoma
histology) of BM development in radically treated stage III NSCLC
patients.
Collapse
Affiliation(s)
- Simon A Keek
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Esma Kayan
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Avishek Chatterjee
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - José S A Belderbos
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Gerben Bootsma
- Department of Pulmonary Diseases, Zuyderland Hospital, Heerlen, The Netherlands
| | - Ben van den Borne
- Department of Pulmonary Diseases, Catharina Hospital, Eindhoven, The Netherlands
| | | | - Hester A Gietema
- Department of Radiology and Nuclear Medicine, GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Harry J M Groen
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Judith Herder
- Department of Pulmonary Diseases, Meander Medical Center, Amersfoort, The Netherlands
| | - Cordula Pitz
- Department of Pulmonary Diseases, Laurentius Hospital, Roermond, The Netherlands
| | - John Praag
- Department of Radiotherapy, Erasmus MC, Rotterdam, The Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (Maastro), GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Janna Schoenmaekers
- Department of Pulmonary Diseases, GROW - School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Hans J M Smit
- Department of Pulmonary Diseases, Rijnstate, Arnhem, The Netherlands
| | - Jos Stigt
- Department of Pulmonary Diseases, Isala Hospital, Zwolle, The Netherlands
| | - Marcel Westenend
- Department of Pulmonary Diseases, VieCuri, Venlo, The Netherlands
| | - Haiyan Zeng
- Department of Radiation Oncology (Maastro), GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Henry C Woodruff
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Lizza Hendriks
- Department of Pulmonary Diseases, GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| |
Collapse
|