1
|
Hull NC, Frush DP, Chu WCW, Ugas Charcape CF, Kasznia-Brown J, Lee EY. Pediatric Imaging 2040. Radiology 2025; 315:e250378. [PMID: 40423538 DOI: 10.1148/radiol.250378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Affiliation(s)
- Nathan C Hull
- Department of Radiology, Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Donald P Frush
- Department of Radiology, Duke University Medical Center, Durham, NC
| | - Winnie C W Chu
- Department of Imaging and Interventional Radiology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Carlos F Ugas Charcape
- Department of Diagnostic Imaging, Instituto Nacional de Salud del Niño San Borja, Lima, Peru
- Now with Pediatric Radiology Section, UH Rainbow Babies and Children's Hospital, Cleveland, Ohio
| | - Joanna Kasznia-Brown
- Department of Radiology, University of Bristol Musgrove Park Hospital, Taunton, United Kingdom
| | - Edward Y Lee
- Department of Radiology, Boston Children's Hospital, Boston, Mass
| |
Collapse
|
2
|
Wang SR, Shen YT, Huang B, Xu HX. Ultrasound-based radiogenomics: status, applications, and future direction. Ultrasonography 2025; 44:95-111. [PMID: 39935290 PMCID: PMC11938802 DOI: 10.14366/usg.24152] [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: 08/12/2024] [Accepted: 12/12/2024] [Indexed: 02/13/2025] Open
Abstract
Radiogenomics, an extension of radiomics, explores the relationship between imaging features and underlying gene expression patterns. This field is instrumental in providing reliable imaging surrogates, thus potentially representing an alternative to genetic testing. The rapidly growing area of radiogenomics that utilizes ultrasound (US) imaging seeks to elucidate the connections between US image characteristics and genomic data. In this review, the authors outline the radiogenomics workflow and summarize the applications of US-based radiogenomics. These include the prediction of gene variations, molecular subtypes, and other biological characteristics, as well as the exploration of the relationships between US phenotypes and cancer gene profiles. Although the field faces various challenges, US-based radiogenomics offers promising prospects and avenues for future research.
Collapse
Affiliation(s)
- Si-Rui Wang
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu-Ting Shen
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Huang
- Department of Ultrasound, Zhejiang Hospital, Hangzhou, China
| | - Hui-Xiong Xu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Liu H, Zhang X, Liu Q. A review of AI-based radiogenomics in neurodegenerative disease. Front Big Data 2025; 8:1515341. [PMID: 40052173 PMCID: PMC11882605 DOI: 10.3389/fdata.2025.1515341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Neurodegenerative diseases are chronic, progressive conditions that cause irreversible damage to the nervous system, particularly in aging populations. Early diagnosis is a critical challenge, as these diseases often develop slowly and without clear symptoms until significant damage has occurred. Recent advances in radiomics and genomics have provided valuable insights into the mechanisms of these diseases by identifying specific imaging features and genomic patterns. Radiogenomics enhances diagnostic capabilities by linking genomics with imaging phenotypes, offering a more comprehensive understanding of disease progression. The growing field of artificial intelligence (AI), including machine learning and deep learning, opens new opportunities for improving the accuracy and timeliness of these diagnoses. This review examines the application of AI-based radiogenomics in neurodegenerative diseases, summarizing key model designs, performance metrics, publicly available data resources, significant findings, and future research directions. It provides a starting point and guidance for those seeking to explore this emerging area of study.
Collapse
Affiliation(s)
- Huanjing Liu
- The Department of Applied Computer Science, Faculty of Science, University of Winnipeg, Winnipeg, MB, Canada
| | - Xiao Zhang
- The Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Qian Liu
- The Department of Applied Computer Science, Faculty of Science, University of Winnipeg, Winnipeg, MB, Canada
- The Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
4
|
Chen W, Gao P, Lu F, Wang E, Liu H, Li M. CT texture features of lung adenocarcinoma with HER2 mutation. BMC Cancer 2025; 25:287. [PMID: 39966778 PMCID: PMC11837593 DOI: 10.1186/s12885-025-13686-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Mutations in human receptor tyrosine kinase epidermal growth factor receptor-2 (HER2) are rare. This study aimed to investigate the clinical characteristics and computed tomography (CT) texture features of lung adenocarcinoma (LUAD) patients with HER2 mutation. METHODS This study included 933 LUAD patients from January 2018 to December 2023 and classified their CT textures accordingly. RESULTS The data indicated that the incidence of HER2 mutation was higher in younger LUAD patients than in elder patients [7.5% (31/413) vs. 1.5% (8/520), p < 0.0001] and was associated with never-smokers [0% (0/78) vs. 4.6% (39/855), p = 0.03]. In this study, the tumors were categorized based on their diameter into T1a and ≥ T1b. The data revealed that HER2 mutation was more frequent in T1a than in ≥ T1b [11.0% (23/210) vs. 2.2% (16/723), p < 0.0001]. Furthermore, non-pSD was more common than pSD in LUAD with HER2 mutation than in LUAD with HER2 wild type [82.1% (31/39) vs.17.9% (7/39), p = 0.01]. Moreover, the size of pGGO (0.94 ± 0.29 cm vs. 1.24 ± 0.39 cm, p = 0.0009), mGGO (0.86 ± 0.39 cm vs. 1.5 ± 0.77 cm, p < 0.0001) and pSD (1.75 ± 0.81 cm vs. 2.5 ± 1.4 cm, p < 0.05) in LUAD patients with HER2 mutation was smaller than those with HER2 wild type patients. In addition, when LUADs with HER2 wild type transformed from pGGO to mGGO, their sizes increased significantly (1.50 ± 0.77 cm vs. 1.24 ± 0.39 cm). It was also observed that the incidence of LUAD with HER2 mutation of ≤ 1 cm was significantly more than that of > 1 cm comparing to that in LUAD with HER2 wild type [14.8% (12/81) vs. 1.7% (3/173), p < 0.0001]. CONCLUSION This study indicated that the incidence of HER2 mutation was higher in younger and never-smoking LUAD patients. Furthermore, the growth of LUAD with HER2 mutation was slower than that of those with HER2 wild type. Moreover, most LUAD with HER2 mutation changed into pSD after > 1 cm.
Collapse
Affiliation(s)
- Wufei Chen
- Department of Radiology, Huadong Hospital, Fudan University Shanghai China, Shanghai, China
| | - Pan Gao
- Department of Radiology, Huadong Hospital, Fudan University Shanghai China, Shanghai, China
| | - Fang Lu
- Department of Radiology, Huadong Hospital, Fudan University Shanghai China, Shanghai, China
| | - Ernuo Wang
- Department of Radiology, Huadong Hospital, Fudan University Shanghai China, Shanghai, China
| | - Haiquan Liu
- Department of Radiology, Huadong Hospital, Fudan University Shanghai China, Shanghai, China.
| | - Ming Li
- Department of Radiology, Huadong Hospital, Fudan University Shanghai China, Shanghai, China.
| |
Collapse
|
5
|
Miyamoto Y, Nakaura T, Ohuchi M, Ogawa K, Kato R, Maeda Y, Eto K, Iwatsuki M, Baba Y, Hirai T, Baba H. Radiomics-based Machine Learning Approach to Predict Chemotherapy Responses in Colorectal Liver Metastases. J Anus Rectum Colon 2025; 9:117-126. [PMID: 39882217 PMCID: PMC11772800 DOI: 10.23922/jarc.2024-077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/15/2024] [Indexed: 01/31/2025] Open
Abstract
Objectives This study explored the clinical utility of CT radiomics-driven machine learning as a predictive marker for chemotherapy response in colorectal liver metastasis (CRLM) patients. Methods We included 150 CRLM patients who underwent first-line doublet chemotherapy, dividing them into a training cohort (n=112) and a test cohort (n=38). We manually delineated three-dimensional tumor volumes, selecting the largest liver metastasis for measurement, using pretreatment portal-phase CT images and extracted 107 radiomics features. Treatment response was classified as responder (complete or partial response) or non-responder (stable or progressive disease), based on the best overall response according to RECIST criteria, version 1.1. Employing Random Forest and Boruta algorithms, we identified significant features for responder-non-responder differentiation. Radiomics signatures were developed and validated in the training cohort using five-fold cross-validation, and performance was assessed using the area under the curve (AUC). Results Among the patients, 91 (61%) were responders and 59 (39%) were non-responders. Variable selection with Boruta revealed three key parameters ("DependenceVariance," "ClusterShade," and "RunVariance"). In the training cohort, individual CT texture parameter AUCs ranged from 0.4 to 0.65, while the machine learning analysis incorporating all valid parameters exhibited a significantly higher AUC of 0.94 (p<0.01). The validation cohort also demonstrated strong predictive accuracy, with an AUC of 0.87 for treatment response. Conclusions This study highlights the potential of CT radiomics-driven machine learning in predicting chemotherapy responses among CRLM patients.
Collapse
Affiliation(s)
- Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Nakaura
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Mayuko Ohuchi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsuhiro Ogawa
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Rikako Kato
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuto Maeda
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kojiro Eto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Division of Translational Research and Advanced Treatment Against Gastrointestinal Cancer, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Next-Generation Surgical Therapy Development, Kumamoto University, Kumamoto, Japan
| | - Toshinori Hirai
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
6
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408069. [PMID: 39535476 PMCID: PMC11727298 DOI: 10.1002/advs.202408069] [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: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Tianxiang Li
- Department of UltrasoundState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical. SciencesPeking Union Medical CollegeBeijing100730China
| | - Bingxin Gong
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and EngineeringSouthern University of Science and TechnologyShenzhen518055China
| | - Sichen Wang
- School of Life Science and TechnologyComputational Biology Research CenterHarbin Institute of TechnologyHarbin150001China
| | - Lian Yang
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Chuansheng Zheng
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| |
Collapse
|
7
|
Okunlola FO, Adetuyi TG, Olajide PA, Okunlola AR, Adetuyi BO, Adeyemo-Eleyode VO, Akomolafe AA, Yunana N, Baba F, Nwachukwu KC, Oyewole OA, Adetunji CO, Shittu OB, Ginikanwa EG. Biomedical image characterization and radio genomics using machine learning techniques. MINING BIOMEDICAL TEXT, IMAGES AND VISUAL FEATURES FOR INFORMATION RETRIEVAL 2025:397-421. [DOI: 10.1016/b978-0-443-15452-2.00019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
8
|
Wang FA, Li Y, Zeng T. Deep Learning of radiology-genomics integration for computational oncology: A mini review. Comput Struct Biotechnol J 2024; 23:2708-2716. [PMID: 39035833 PMCID: PMC11260400 DOI: 10.1016/j.csbj.2024.06.019] [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: 03/06/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
In the field of computational oncology, patient status is often assessed using radiology-genomics, which includes two key technologies and data, such as radiology and genomics. Recent advances in deep learning have facilitated the integration of radiology-genomics data, and even new omics data, significantly improving the robustness and accuracy of clinical predictions. These factors are driving artificial intelligence (AI) closer to practical clinical applications. In particular, deep learning models are crucial in identifying new radiology-genomics biomarkers and therapeutic targets, supported by explainable AI (xAI) methods. This review focuses on recent developments in deep learning for radiology-genomics integration, highlights current challenges, and outlines some research directions for multimodal integration and biomarker discovery of radiology-genomics or radiology-omics that are urgently needed in computational oncology.
Collapse
Affiliation(s)
- Feng-ao Wang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yixue Li
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Guangzhou National Laboratory, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Tao Zeng
- Guangzhou National Laboratory, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
9
|
Pecchi A, Bozzola C, Beretta C, Besutti G, Toss A, Cortesi L, Balboni E, Nocetti L, Ligabue G, Torricelli P. DCE-MRI Radiomic analysis in triple negative ductal invasive breast cancer. Comparison between BRCA and not BRCA mutated patients: Preliminary results. Magn Reson Imaging 2024; 113:110214. [PMID: 39047852 DOI: 10.1016/j.mri.2024.110214] [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: 05/13/2024] [Revised: 07/17/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE The research aimed to determine whether and which radiomic features from breast dynamic contrast enhanced (DCE) MRI could predict the presence of BRCA1 mutation in patients with triple-negative breast cancer (TNBC). MATERIAL AND METHODS This retrospective study included consecutive patients histologically diagnosed with TNBC who underwent breast DCE-MRI in 2010-2021. Baseline DCE-MRIs were retrospectively reviewed; percentage maps of wash-in and wash-out were computed and breast lesions were manually segmented, drawing a 5 mm-Region of Interest (ROI) inside the tumor and another 5 mm-ROI inside the contralateral healthy gland. Features for each map and each ROI were extracted with Pyradiomics-3D Slicer and considered first separately (tumor and contralateral gland) and then together. In each analysis the more important features for BRCA1 status classification were selected with Maximum Relevance Minimum Redundancy algorithm and used to fit four classifiers. RESULTS The population included 67 patients and 86 lesions (21 in BRCA1-mutated, 65 in non BRCA-carriers). The best classifiers for BRCA mutation were Support Vector Classifier and Logistic Regression in models fitted with both gland and tumor features, reaching an Area Under ROC Curve (AUC) of 0.80 (SD 0.21) and of 0.79 (SD 0.20), respectively. Three features were higher in BRCA1-mutated compared to non BRCA-mutated: Total Energy and Correlation from gray level cooccurrence matrix, both measured in contralateral gland in wash-out maps, and Root Mean Squared, selected from the wash-out map of the tumor. CONCLUSIONS This study showed the feasibility of a radiomic study with breast DCE-MRI and the potential of radiomics in predicting BRCA1 mutational status.
Collapse
Affiliation(s)
- Annarita Pecchi
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Chiara Bozzola
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Cecilia Beretta
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Giulia Besutti
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy; Radiology Unit, Department of Diagnostic Imaging and Laboratory Medicine, AUSL-IRCCS di Reggio Emilia, Via Risorgimento 80, 42123 Reggio Emilia, Italy.
| | - Angela Toss
- Division of Oncology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Laura Cortesi
- Division of Oncology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Erica Balboni
- Medical Physics Unit, University Hospital of Modena, 41124 Modena, Italy
| | - Luca Nocetti
- Medical Physics Unit, University Hospital of Modena, 41124 Modena, Italy
| | - Guido Ligabue
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| | - Pietro Torricelli
- Division of Radiology, Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41224 Modena, Italy
| |
Collapse
|
10
|
Sha X, Wang C, Qi S, Yuan X, Zhang H, Yang J. The efficacy of CBCT-based radiomics techniques in differentiating between conventional and unicystic ameloblastoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2024; 138:656-665. [PMID: 39227265 DOI: 10.1016/j.oooo.2024.06.010] [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: 02/07/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 09/05/2024]
Abstract
OBJECTIVE The aim of this study was to develop a cone beam computed tomography (CBCT) radiomics-based model that differentiates between conventional and unicystic ameloblastoma (AB). METHODS In this retrospective study, CBCT images were collected from 100 patients who had ABs that were diagnosed histopathologically as conventional or unicystic AB after surgical treatment. The patients were randomly divided into training (70) and validation (30) cohorts. Radiomics features were extracted from the images, and the optimal features were incorporated into 5 models: Logistic Regression, Support Vector Machine, Linear Discriminant Analysis, Random Forest, and XGBoost for prediction of tumor type. Model performance was evaluated using the area under the curve (AUC) from receiver operating characteristic analysis, sensitivity, specificity, accuracy, calibration curves, and decision curve analysis (DCA). RESULTS The 20 optimal radiomics features were incorporated into the Logistic Regression (LR) model, which exhibited the best overall performance with AUC = 0.936 (95% confidence interval [CI] = 0.877-0.996) for the training cohort and AUC = 0.929 (95% CI = 0.832-1.000) for the validation cohort. The nomogram combined the clinical features and the radiomics signature and resulted in the best predictive performance. CONCLUSIONS The LR model demonstrated the ability of radiomics and the nomogram to distinguish between the 2 types of AB and may have the potential to replace biopsies under noninvasive conditions.
Collapse
Affiliation(s)
- Xiaoyan Sha
- Department of Oral and Maxillofacial Radiology, School of Stomatology, Capital Medical University, Beijing, China
| | - Chao Wang
- Department of Clinical Research, SinoUnion Healthcare Inc., Beijing, China
| | - Senrong Qi
- Department of Oral and Maxillofacial Radiology, School of Stomatology, Capital Medical University, Beijing, China
| | - Xiaohong Yuan
- Department of Oral and Maxillofacial Pathology, School of Stomatology, Capital Medical University, Beijing, China
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
11
|
Jin Z, Zou Q, Zhou T, Xue T. Preoperative prediction of early recurrence in patients with BRAF mutant colorectal cancer using a intergrated nomogram. Sci Rep 2024; 14:25320. [PMID: 39455810 PMCID: PMC11512039 DOI: 10.1038/s41598-024-77256-2] [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: 05/23/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024] Open
Abstract
Objective To explore the predictive value of radiomics nomogram combining with CT radiomics features and clinical features for postoperative early recurrence in patients with BRAF-mutant colorectal cancer. Methods A total of 220 patients with surgically and pathologically confirmed BRAF-mutant colorectal cancer from 2 institutions were retrospectively included. All patients from institution 1 were randomized at a 7:3 ratio into a training cohort (n = 108) and an internal validation cohort (n = 45), and patients from institution 2 were used as an external validation cohort (n = 67). The association between the radiomics features and early recurrence was assessed in the training cohort and verified in the validation cohort. Furthermore, the performance of the radiomics nomogram was evaluated by combining the rad-score and clinical risk factors. The predictive performance was evaluated by receiver operating characteristic curve analysis, calibration curve analysis, and decision curve analysis. Results The dierenees in the Lymphocyte/monocyte ratio (LMR) and Peripheral nerve infiltration (PNI) were statistically significant between the early recurrence in BRAF-mutant colorectal cancer groups and the early non-recurrence in BRAF-mutant colorectal cancer groups (P < 0.05); The two groups showed no significant differenee in clinical parameters including age, sex, and biochemistry serum markers (P > 0.05). Comparing with the pure radiomics or clinical data, combined models can be seen that the addition of LMR and PNI further improveed the predictive efficiency of the model. The rad-score based on LR, generated by 4 selected radiomics features, demonstrated a favorable ability to predict early recurrence in both the training (AUC 0.81), internal validation (AUC 0.73), external validation (AUC 0.63) cohorts. Subsequently, integrating two independent predictors into a nomogram exhibited more favorable discriminatory performance, with the AUC improved to 0.88 and 0.81 in both cohorts. Conclusions The proposed CT-based radiomics signature is associated with early recurrence among the patients with BRAF-mutant colorectal cancer. The present study also proposes a combined model can potentially be applied in the individual preoperative prediction of early recurrence in patients with BRAF-mutant colorectal cancer. Advances in knowledge CT-based radiomics showed satisfactory diagnostic significance for early recurrence in patients with BRAF-mutant colorectal cancer. Key baseline clinical characteristics were associated with early recurrence in patients with BRAF-mutant colorectal cancer. The combined model may be applied in the individual preoperative prediction of early recurrence in patients with BRAF-mutant colorectal cancer.
Collapse
Affiliation(s)
- Zhenbin Jin
- Department of Radiology, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, #60 Youth Middle Road, Chongchuan District, Nantong, 226000, Jiangsu, China
| | - Qin Zou
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Taohu Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Ting Xue
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
12
|
Chen L, Huang ZH, Sun Y, Domaratzki M, Liu Q, Hu P. Conditional probabilistic diffusion model driven synthetic radiogenomic applications in breast cancer. PLoS Comput Biol 2024; 20:e1012490. [PMID: 39374308 PMCID: PMC11486376 DOI: 10.1371/journal.pcbi.1012490] [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: 01/25/2024] [Revised: 10/17/2024] [Accepted: 09/14/2024] [Indexed: 10/09/2024] Open
Abstract
This study addresses the heterogeneity of Breast Cancer (BC) by employing a Conditional Probabilistic Diffusion Model (CPDM) to synthesize Magnetic Resonance Images (MRIs) based on multi-omic data, including gene expression, copy number variation, and DNA methylation. The lack of paired medical images and genomics data in previous studies presented a challenge, which the CPDM aims to overcome. The well-trained CPDM successfully generated synthetic MRIs for 726 TCGA-BRCA patients, who lacked actual MRIs, using their multi-omic profiles. Evaluation metrics such as Frechet's Inception Distance (FID), Mean Square Error (MSE), and Structural Similarity Index Measure (SSIM) demonstrated the CPDM's effectiveness, with an FID of 2.02, an MSE of 0.02, and an SSIM of 0.59 based on the 15-fold cross-validation. The synthetic MRIs were used to predict clinical attributes, achieving an Area Under the Receiver-Operating-Characteristic curve (AUROC) of 0.82 and an Area Under the Precision-Recall Curve (AUPRC) of 0.84 for predicting ER+/HER2+ subtypes. Additionally, the MRIs served to accurately predicted BC patient survival with a Concordance-index (C-index) score of 0.88, outperforming other baseline models. This research demonstrates the potential of CPDMs in generating MRIs based on BC patients' genomic profiles, offering valuable insights for radiogenomic research and advancements in precision medicine. The study provides a novel approach to understanding BC heterogeneity for early detection and personalized treatment.
Collapse
Affiliation(s)
- Lianghong Chen
- Department of Computer Science, Western University, London, Ontario, Canada
| | - Zi Huai Huang
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Yan Sun
- Department of Computer Science, Western University, London, Ontario, Canada
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mike Domaratzki
- Department of Computer Science, Western University, London, Ontario, Canada
| | - Qian Liu
- Department of Applied Computer Science, University of Winnipeg, Winnipeg, Manitoba, Canada
| | - Pingzhao Hu
- Department of Computer Science, Western University, London, Ontario, Canada
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- The Children’s Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
13
|
Chen J, Zeng H, Cheng Y, Yang B. Identifying radiogenomic associations of breast cancer based on DCE-MRI by using Siamese Neural Network with manufacturer bias normalization. Med Phys 2024; 51:7269-7281. [PMID: 38922986 DOI: 10.1002/mp.17266] [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: 01/17/2024] [Revised: 06/08/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND AND PURPOSE The immunohistochemical test (IHC) for Human Epidermal Growth Factor Receptor 2 (HER2) and hormone receptors (HR) provides prognostic information and guides treatment for patients with invasive breast cancer. The objective of this paper is to establish a non-invasive system for identifying HER2 and HR in breast cancer using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). METHODS In light of the absence of high-performance algorithms and external validation in previously published methods, this study utilizes 3D deep features and radiomics features to represent the information of the Region of Interest (ROI). A Siamese Neural Network was employed as the classifier, with 3D deep features and radiomics features serving as the network input. To neutralize manufacturer bias, a batch effect normalization method, ComBat, was introduced. To enhance the reliability of the study, two datasets, Predict Your Therapeutic Response with Imaging and moLecular Analysis (I-SPY 1) and I-SPY 2, were incorporated. I-SPY 2 was utilized for model training and validation, while I-SPY 1 was exclusively employed for external validation. Additionally, a breast tumor segmentation network was trained to improve radiomic feature extraction. RESULTS The results indicate that our approach achieved an average Area Under the Curve (AUC) of 0.632, with a Standard Error of the Mean (SEM) of 0.042 for HER2 prediction in the I-SPY 2 dataset. For HR prediction, our method attained an AUC of 0.635 (SEM 0.041), surpassing other published methods in the AUC metric. Moreover, the proposed method yielded competitive results in other metrics. In external validation using the I-SPY 1 dataset, our approach achieved an AUC of 0.567 (SEM 0.032) for HR prediction and 0.563 (SEM 0.033) for HER2 prediction. CONCLUSION This study proposes a non-invasive system for identifying HER2 and HR in breast cancer. Although the results do not conclusively demonstrate superiority in both tasks, they indicate that the proposed method achieved good performance and is a competitive classifier compared to other reference methods. Ablation studies demonstrate that both radiomics features and deep features for the Siamese Neural Network are beneficial for the model. The introduced manufacturer bias normalization method has been shown to enhance the method's performance. Furthermore, the external validation of the method enhances the reliability of this research. Source code, pre-trained segmentation network, Radiomics and deep features, data for statistical analysis, and Supporting Information of this article are online at: https://github.com/FORRESTHUACHEN/Siamese_Neural_Network_based_Brest_cancer_Radiogenomic.
Collapse
Affiliation(s)
- Junhua Chen
- School of Medicine, Shanghai University, Shanghai, China
| | - Haiyan Zeng
- Department of Radiation Oncology, Division of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yanyan Cheng
- Medical Engineering Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, China
| | - Banghua Yang
- School of Medicine, Shanghai University, Shanghai, China
- School of Mechatronic Engineering and Automation, Research Center of Brain Computer Engineering, Shanghai University, Shanghai, China
| |
Collapse
|
14
|
Chen W, Zhang W, Chen X, Dong W, Cai Y, Cheng J, Jin J. Computed tomography-based radiomics nomogram for predicting therapeutic response to neoadjuvant chemotherapy in locally advanced gastric cancer : A scale for treatment predicting. Clin Transl Oncol 2024; 26:1944-1955. [PMID: 38467894 DOI: 10.1007/s12094-024-03417-4] [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: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND AND OBJECTIVE Neoadjuvant chemotherapy results in various responses when used to treat locally advanced gastric cancer, we aimed to develop and validate a predictive model of the response to neoadjuvant chemotherapy in patients with gastric cancer. METHODS A total of 128 patients with locally advanced gastric cancer who underwent pre-treatment computed tomography (CT) scanning followed by neoadjuvant chemoradiotherapy were included (training cohort: n = 64; validation cohort: n = 64). We built a radiomics score combined with laboratory parameters to create a nomogram for predicting the efficacy of neoadjuvant chemotherapy and calculating scores for risk factors. RESULTS The radiomics score system demonstrated good stability and prediction performance for the response to neoadjuvant chemotherapy, with the area under the curve of the training and validation cohorts being 0.8 and 0.64, respectively. The radiomics score proved to be an independent risk factor affecting the efficacy of neoadjuvant chemotherapy. In addition to the radiomics score, four other risk factors were included in the nomogram, namely the platelet-to-lymphocyte ratio, total bilirubin, ALT/AST, and CA199. The model had a C-index of 0.8. CONCLUSIONS Our results indicated that radiomics features could be potential biomarkers for the early prediction of the response to neoadjuvant treatment.
Collapse
Affiliation(s)
- Wenjing Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiteng Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xietao Chen
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weisong Dong
- Department of Pathology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiqi Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jinji Jin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
15
|
Wang M, Peng Y, Wang Y, Luo D. Research Trends and Evolution in Radiogenomics (2005-2023): Bibliometric Analysis. Interact J Med Res 2024; 13:e51347. [PMID: 38980713 PMCID: PMC11267093 DOI: 10.2196/51347] [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: 07/28/2023] [Revised: 03/10/2024] [Accepted: 05/20/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Radiogenomics is an emerging technology that integrates genomics and medical image-based radiomics, which is considered a promising approach toward achieving precision medicine. OBJECTIVE The aim of this study was to quantitatively analyze the research status, dynamic trends, and evolutionary trajectory in the radiogenomics field using bibliometric methods. METHODS The relevant literature published up to 2023 was retrieved from the Web of Science Core Collection. Excel was used to analyze the annual publication trend. VOSviewer was used for constructing the keywords co-occurrence network and the collaboration networks among countries and institutions. CiteSpace was used for citation keywords burst analysis and visualizing the references timeline. RESULTS A total of 3237 papers were included and exported in plain-text format. The annual number of publications showed an increasing annual trend. China and the United States have published the most papers in this field, with the highest number of citations in the United States and the highest average number per item in the Netherlands. Keywords burst analysis revealed that several keywords, including "big data," "magnetic resonance spectroscopy," "renal cell carcinoma," "stage," and "temozolomide," experienced a citation burst in recent years. The timeline views demonstrated that the references can be categorized into 8 clusters: lower-grade glioma, lung cancer histology, lung adenocarcinoma, breast cancer, radiation-induced lung injury, epidermal growth factor receptor mutation, late radiotherapy toxicity, and artificial intelligence. CONCLUSIONS The field of radiogenomics is attracting increasing attention from researchers worldwide, with the United States and the Netherlands being the most influential countries. Exploration of artificial intelligence methods based on big data to predict the response of tumors to various treatment methods represents a hot spot research topic in this field at present.
Collapse
Affiliation(s)
- Meng Wang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yun Peng
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Ya Wang
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dehong Luo
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| |
Collapse
|
16
|
Dragon JM, Guha S, Salvatore MM. Hounsfield units: Future applications in clinical practice, radiomics, and Artificial Intelligence. Clin Imaging 2024; 110:110141. [PMID: 38608412 DOI: 10.1016/j.clinimag.2024.110141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/21/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024]
Affiliation(s)
- Jacqueline M Dragon
- Department of Radiology, Columbia University Vagelos College of Physicians and Surgeons, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, NY, United States of America
| | - Siddharth Guha
- Department of Radiology, Columbia University Vagelos College of Physicians and Surgeons, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, NY, United States of America
| | - Mary M Salvatore
- Department of Radiology, Columbia University Vagelos College of Physicians and Surgeons, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, NY, United States of America.
| |
Collapse
|
17
|
Xue T, Wan X, Zhou T, Zou Q, Ma C, Chen J. Potential value of CT-based comprehensive nomogram in predicting occult lymph node metastasis of esophageal squamous cell paralaryngeal nerves: a two-center study. J Transl Med 2024; 22:399. [PMID: 38689366 PMCID: PMC11059581 DOI: 10.1186/s12967-024-05217-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024] Open
Abstract
PURPOSE The aim of this study is to construct a combined model that integrates radiomics, clinical risk factors and machine learning algorithms to predict para-laryngeal lymph node metastasis in esophageal squamous cell carcinoma. METHODS A retrospective study included 361 patients with esophageal squamous cell carcinoma from 2 centers. Radiomics features were extracted from the computed tomography scans. Logistic regression, k nearest neighbor, multilayer perceptron, light Gradient Boosting Machine, support vector machine, random forest algorithms were used to construct radiomics models. The receiver operating characteristic curve and The Hosmer-Lemeshow test were employed to select the better-performing model. Clinical risk factors were identified through univariate logistic regression analysis and multivariate logistic regression analysis and utilized to develop a clinical model. A combined model was then created by merging radiomics and clinical risk factors. The performance of the models was evaluated using ROC curve analysis, and the clinical value of the models was assessed using decision curve analysis. RESULTS A total of 1024 radiomics features were extracted. Among the radiomics models, the KNN model demonstrated the optimal diagnostic capabilities and accuracy, with an area under the curve (AUC) of 0.84 in the training cohort and 0.62 in the internal test cohort. Furthermore, the combined model exhibited an AUC of 0.97 in the training cohort and 0.86 in the internal test cohort. CONCLUSION A clinical-radiomics integrated nomogram can predict occult para-laryngeal lymph node metastasis in esophageal squamous cell carcinoma and provide guidance for personalized treatment.
Collapse
Affiliation(s)
- Ting Xue
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China.
| | - Xinyi Wan
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Taohu Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Qin Zou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Chao Ma
- Department of Radiology, Frist Affiliated Hospital of Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
| | - Jieqiong Chen
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| |
Collapse
|
18
|
Khodabakhshi Z, Motisi L, Bink A, Broglie MA, Rupp NJ, Fleischmann M, von der Grün J, Guckenberger M, Tanadini-Lang S, Balermpas P. MRI-based radiomics for predicting histology in malignant salivary gland tumors: methodology and "proof of principle". Sci Rep 2024; 14:9945. [PMID: 38688932 PMCID: PMC11061101 DOI: 10.1038/s41598-024-60200-9] [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: 12/01/2023] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
Defining the exact histological features of salivary gland malignancies before treatment remains an unsolved problem that compromises the ability to tailor further therapeutic steps individually. Radiomics, a new methodology to extract quantitative information from medical images, could contribute to characterizing the individual cancer phenotype already before treatment in a fast and non-invasive way. Consequently, the standardization and implementation of radiomic analysis in the clinical routine work to predict histology of salivary gland cancer (SGC) could also provide improvements in clinical decision-making. In this study, we aimed to investigate the potential of radiomic features as imaging biomarker to distinguish between high grade and low-grade salivary gland malignancies. We have also investigated the effect of image and feature level harmonization on the performance of radiomic models. For this study, our dual center cohort consisted of 126 patients, with histologically proven SGC, who underwent curative-intent treatment in two tertiary oncology centers. We extracted and analyzed the radiomics features of 120 pre-therapeutic MRI images with gadolinium (T1 sequences), and correlated those with the definitive post-operative histology. In our study the best radiomic model achieved average AUC of 0.66 and balanced accuracy of 0.63. According to the results, there is significant difference between the performance of models based on MRI intensity normalized images + harmonized features and other models (p value < 0.05) which indicates that in case of dealing with heterogeneous dataset, applying the harmonization methods is beneficial. Among radiomic features minimum intensity from first order, and gray level-variance from texture category were frequently selected during multivariate analysis which indicate the potential of these features as being used as imaging biomarker. The present bicentric study presents for the first time the feasibility of implementing MR-based, handcrafted radiomics, based on T1 contrast-enhanced sequences and the ComBat harmonization method in an effort to predict the formal grading of salivary gland carcinoma with satisfactory performance.
Collapse
Affiliation(s)
- Zahra Khodabakhshi
- Department of Radiation Oncology, Zurich University Hospital, Zurich, Switzerland
| | - Laura Motisi
- Department of Radiation Oncology, Zurich University Hospital, Zurich, Switzerland
| | - Andrea Bink
- Department of Neuroradadiology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Martina A Broglie
- Department of Otorhinolaryngology, Zurich University Hospital, Zurich, Switzerland
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Maximilian Fleischmann
- Department of Radiation Oncology, J.W. Goethe University Hospital Frankfurt, Frankfurt, Germany
| | - Jens von der Grün
- Department of Radiation Oncology, Zurich University Hospital, Zurich, Switzerland
| | | | | | - Panagiotis Balermpas
- Department of Radiation Oncology, Zurich University Hospital, Zurich, Switzerland.
| |
Collapse
|
19
|
Santoro M, Zybin V, Coada CA, Mantovani G, Paolani G, Di Stanislao M, Modolon C, Di Costanzo S, Lebovici A, Ravegnini G, De Leo A, Tesei M, Pasquini P, Lovato L, Morganti AG, Pantaleo MA, De Iaco P, Strigari L, Perrone AM. Machine Learning Applied to Pre-Operative Computed-Tomography-Based Radiomic Features Can Accurately Differentiate Uterine Leiomyoma from Leiomyosarcoma: A Pilot Study. Cancers (Basel) 2024; 16:1570. [PMID: 38672651 PMCID: PMC11048510 DOI: 10.3390/cancers16081570] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The accurate discrimination of uterine leiomyosarcomas and leiomyomas in a pre-operative setting remains a current challenge. To date, the diagnosis is made by a pathologist on the excised tumor. The aim of this study was to develop a machine learning algorithm using radiomic data extracted from contrast-enhanced computed tomography (CECT) images that could accurately distinguish leiomyosarcomas from leiomyomas. METHODS Pre-operative CECT images from patients submitted to surgery with a histological diagnosis of leiomyoma or leiomyosarcoma were used for the region of interest identification and radiomic feature extraction. Feature extraction was conducted using the PyRadiomics library, and three feature selection methods combined with the general linear model (GLM), random forest (RF), and support vector machine (SVM) classifiers were built, trained, and tested for the binary classification task (malignant vs. benign). In parallel, radiologists assessed the diagnosis with or without clinical data. RESULTS A total of 30 patients with leiomyosarcoma (mean age 59 years) and 35 patients with leiomyoma (mean age 48 years) were included in the study, comprising 30 and 51 lesions, respectively. Out of nine machine learning models, the three feature selection methods combined with the GLM and RF classifiers showed good performances, with predicted area under the curve (AUC), sensitivity, and specificity ranging from 0.78 to 0.97, from 0.78 to 1.00, and from 0.67 to 0.93, respectively, when compared to the results obtained from experienced radiologists when blinded to the clinical profile (AUC = 0.73 95%CI = 0.62-0.84), as well as when the clinical data were consulted (AUC = 0.75 95%CI = 0.65-0.85). CONCLUSIONS CECT images integrated with radiomics have great potential in differentiating uterine leiomyomas from leiomyosarcomas. Such a tool can be used to mitigate the risks of eventual surgical spread in the case of leiomyosarcoma and allow for safer fertility-sparing treatment in patients with benign uterine lesions.
Collapse
Affiliation(s)
- Miriam Santoro
- Department of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.S.); (G.P.); (L.S.)
| | - Vladislav Zybin
- Pediatric and Adult CardioThoracic and Vascular, Oncohematologic and Emergency Radiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (V.Z.); (C.M.); (L.L.)
| | | | - Giulia Mantovani
- Division of Oncologic Gynecology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (G.M.); (M.D.S.); (S.D.C.); (M.T.); (P.P.)
| | - Giulia Paolani
- Department of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.S.); (G.P.); (L.S.)
| | - Marco Di Stanislao
- Division of Oncologic Gynecology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (G.M.); (M.D.S.); (S.D.C.); (M.T.); (P.P.)
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.D.L.); (A.G.M.); (M.A.P.)
| | - Cecilia Modolon
- Pediatric and Adult CardioThoracic and Vascular, Oncohematologic and Emergency Radiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (V.Z.); (C.M.); (L.L.)
| | - Stella Di Costanzo
- Division of Oncologic Gynecology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (G.M.); (M.D.S.); (S.D.C.); (M.T.); (P.P.)
| | - Andrei Lebovici
- Radiology and Imaging Department, County Emergency Hospital, 400347 Cluj-Napoca, Romania;
- Surgical Specialties Department, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| | - Antonio De Leo
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.D.L.); (A.G.M.); (M.A.P.)
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Marco Tesei
- Division of Oncologic Gynecology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (G.M.); (M.D.S.); (S.D.C.); (M.T.); (P.P.)
| | - Pietro Pasquini
- Division of Oncologic Gynecology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (G.M.); (M.D.S.); (S.D.C.); (M.T.); (P.P.)
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.D.L.); (A.G.M.); (M.A.P.)
| | - Luigi Lovato
- Pediatric and Adult CardioThoracic and Vascular, Oncohematologic and Emergency Radiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (V.Z.); (C.M.); (L.L.)
| | - Alessio Giuseppe Morganti
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.D.L.); (A.G.M.); (M.A.P.)
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Maria Abbondanza Pantaleo
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.D.L.); (A.G.M.); (M.A.P.)
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Pierandrea De Iaco
- Division of Oncologic Gynecology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (G.M.); (M.D.S.); (S.D.C.); (M.T.); (P.P.)
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.D.L.); (A.G.M.); (M.A.P.)
| | - Lidia Strigari
- Department of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.S.); (G.P.); (L.S.)
| | - Anna Myriam Perrone
- Division of Oncologic Gynecology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (G.M.); (M.D.S.); (S.D.C.); (M.T.); (P.P.)
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.D.L.); (A.G.M.); (M.A.P.)
| |
Collapse
|
20
|
Coroller T, Sahiner B, Amatya A, Gossmann A, Karagiannis K, Moloney C, Samala RK, Santana-Quintero L, Solovieff N, Wang C, Amiri-Kordestani L, Cao Q, Cha KH, Charlab R, Cross FH, Hu T, Huang R, Kraft J, Krusche P, Li Y, Li Z, Mazo I, Paul R, Schnakenberg S, Serra P, Smith S, Song C, Su F, Tiwari M, Vechery C, Xiong X, Zarate JP, Zhu H, Chakravartty A, Liu Q, Ohlssen D, Petrick N, Schneider JA, Walderhaug M, Zuber E. Methodology for Good Machine Learning with Multi-Omics Data. Clin Pharmacol Ther 2024; 115:745-757. [PMID: 37965805 DOI: 10.1002/cpt.3105] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023]
Abstract
In 2020, Novartis Pharmaceuticals Corporation and the U.S. Food and Drug Administration (FDA) started a 4-year scientific collaboration to approach complex new data modalities and advanced analytics. The scientific question was to find novel radio-genomics-based prognostic and predictive factors for HR+/HER- metastatic breast cancer under a Research Collaboration Agreement. This collaboration has been providing valuable insights to help successfully implement future scientific projects, particularly using artificial intelligence and machine learning. This tutorial aims to provide tangible guidelines for a multi-omics project that includes multidisciplinary expert teams, spanning across different institutions. We cover key ideas, such as "maintaining effective communication" and "following good data science practices," followed by the four steps of exploratory projects, namely (1) plan, (2) design, (3) develop, and (4) disseminate. We break each step into smaller concepts with strategies for implementation and provide illustrations from our collaboration to further give the readers actionable guidance.
Collapse
Affiliation(s)
| | - Berkman Sahiner
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Anup Amatya
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Alexej Gossmann
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Konstantinos Karagiannis
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Ravi K Samala
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Luis Santana-Quintero
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Nadia Solovieff
- Novartis Pharmaceutical Company, East Hanover, New Jersey, USA
| | - Craig Wang
- Novartis Pharma AG, Rotkreuz, Switzerland
| | - Laleh Amiri-Kordestani
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Qian Cao
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Kenny H Cha
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Rosane Charlab
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Frank H Cross
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Tingting Hu
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ruihao Huang
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jeffrey Kraft
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Yutong Li
- Novartis Pharmaceutical Company, East Hanover, New Jersey, USA
| | - Zheng Li
- Novartis Pharmaceutical Company, East Hanover, New Jersey, USA
| | - Ilya Mazo
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Rahul Paul
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Paolo Serra
- Novartis Pharmaceutical Company, East Hanover, New Jersey, USA
| | - Sean Smith
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Chi Song
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Fei Su
- Novartis Pharmaceutical Company, East Hanover, New Jersey, USA
| | - Mohit Tiwari
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Colin Vechery
- Novartis Pharmaceutical Company, East Hanover, New Jersey, USA
| | - Xin Xiong
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Hao Zhu
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Qi Liu
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - David Ohlssen
- Novartis Pharmaceutical Company, East Hanover, New Jersey, USA
| | - Nicholas Petrick
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Julie A Schneider
- Oncology Center of Excellence, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mark Walderhaug
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | | |
Collapse
|
21
|
Demetriou D, Lockhat Z, Brzozowski L, Saini KS, Dlamini Z, Hull R. The Convergence of Radiology and Genomics: Advancing Breast Cancer Diagnosis with Radiogenomics. Cancers (Basel) 2024; 16:1076. [PMID: 38473432 DOI: 10.3390/cancers16051076] [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: 01/12/2024] [Revised: 02/09/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Despite significant progress in the prevention, screening, diagnosis, prognosis, and therapy of breast cancer (BC), it remains a highly prevalent and life-threatening disease affecting millions worldwide. Molecular subtyping of BC is crucial for predictive and prognostic purposes due to the diverse clinical behaviors observed across various types. The molecular heterogeneity of BC poses uncertainties in its impact on diagnosis, prognosis, and treatment. Numerous studies have highlighted genetic and environmental differences between patients from different geographic regions, emphasizing the need for localized research. International studies have revealed that patients with African heritage are often diagnosed at a more advanced stage and exhibit poorer responses to treatment and lower survival rates. Despite these global findings, there is a dearth of in-depth studies focusing on communities in the African region. Early diagnosis and timely treatment are paramount to improving survival rates. In this context, radiogenomics emerges as a promising field within precision medicine. By associating genetic patterns with image attributes or features, radiogenomics has the potential to significantly improve early detection, prognosis, and diagnosis. It can provide valuable insights into potential treatment options and predict the likelihood of survival, progression, and relapse. Radiogenomics allows for visual features and genetic marker linkage that promises to eliminate the need for biopsy and sequencing. The application of radiogenomics not only contributes to advancing precision oncology and individualized patient treatment but also streamlines clinical workflows. This review aims to delve into the theoretical underpinnings of radiogenomics and explore its practical applications in the diagnosis, management, and treatment of BC and to put radiogenomics on a path towards fully integrated diagnostics.
Collapse
Affiliation(s)
- Demetra Demetriou
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Zarina Lockhat
- Department of Radiology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Luke Brzozowski
- Translational Research and Core Facilities, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Kamal S Saini
- Fortrea Inc., 8 Moore Drive, Durham, NC 27709, USA
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| |
Collapse
|
22
|
Barat M, Pellat A, Hoeffel C, Dohan A, Coriat R, Fishman EK, Nougaret S, Chu L, Soyer P. CT and MRI of abdominal cancers: current trends and perspectives in the era of radiomics and artificial intelligence. Jpn J Radiol 2024; 42:246-260. [PMID: 37926780 DOI: 10.1007/s11604-023-01504-0] [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: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023]
Abstract
Abdominal cancers continue to pose daily challenges to clinicians, radiologists and researchers. These challenges are faced at each stage of abdominal cancer management, including early detection, accurate characterization, precise assessment of tumor spread, preoperative planning when surgery is anticipated, prediction of tumor aggressiveness, response to therapy, and detection of recurrence. Technical advances in medical imaging, often in combination with imaging biomarkers, show great promise in addressing such challenges. Information extracted from imaging datasets owing to the application of radiomics can be used to further improve the diagnostic capabilities of imaging. However, the analysis of the huge amount of data provided by these advances is a difficult task in daily practice. Artificial intelligence has the potential to help radiologists in all these challenges. Notably, the applications of AI in the field of abdominal cancers are expanding and now include diverse approaches for cancer detection, diagnosis and classification, genomics and detection of genetic alterations, analysis of tumor microenvironment, identification of predictive biomarkers and follow-up. However, AI currently has some limitations that need further refinement for implementation in the clinical setting. This review article sums up recent advances in imaging of abdominal cancers in the field of image/data acquisition, tumor detection, tumor characterization, prognosis, and treatment response evaluation.
Collapse
Affiliation(s)
- Maxime Barat
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hopitaux de Paris, 75014, Paris, France
- Faculté de Médecine, Université Paris Cité, 75006, Paris, France
| | - Anna Pellat
- Faculté de Médecine, Université Paris Cité, 75006, Paris, France
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, Assistance Publique-Hopitaux de Paris, 75014, Paris, France
| | - Christine Hoeffel
- Department of Radiology, Hopital Robert Debré, CHU Reims, Université Champagne-Ardennes, 51092, Reims, France
| | - Anthony Dohan
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hopitaux de Paris, 75014, Paris, France
- Faculté de Médecine, Université Paris Cité, 75006, Paris, France
| | - Romain Coriat
- Faculté de Médecine, Université Paris Cité, 75006, Paris, France
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, Assistance Publique-Hopitaux de Paris, 75014, Paris, France
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Stéphanie Nougaret
- Department of Radiology, Montpellier Cancer Institute, 34000, Montpellier, France
- PINKCC Lab, IRCM, U1194, 34000, Montpellier, France
| | - Linda Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Philippe Soyer
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hopitaux de Paris, 75014, Paris, France.
- Faculté de Médecine, Université Paris Cité, 75006, Paris, France.
| |
Collapse
|
23
|
Qian H, Ren X, Xu M, Fang Z, Zhang R, Bu Y, Zhou C. Magnetic resonance imaging-based radiomics was used to evaluate the level of prognosis-related immune cell infiltration in breast cancer tumor microenvironment. BMC Med Imaging 2024; 24:31. [PMID: 38308230 PMCID: PMC10835863 DOI: 10.1186/s12880-024-01212-9] [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: 02/23/2023] [Accepted: 01/24/2024] [Indexed: 02/04/2024] Open
Abstract
PURPOSE The tumor immune microenvironment is a valuable source of information for predicting prognosis in breast cancer (BRCA) patients. To identify immune cells associated with BRCA patient prognosis from the Cancer Genetic Atlas (TCGA), we established an MRI-based radiomics model for evaluating the degree of immune cell infiltration in breast cancer patients. METHODS CIBERSORT was utilized to evaluate the degree of infiltration of 22 immune cell types in breast cancer patients from the TCGA database, and both univariate and multivariate Cox regressions were employed to determine the prognostic significance of immune cell infiltration levels in BRCA patients. We identified independent prognostic factors for BRCA patients. Additionally, we obtained imaging features from the Cancer Imaging Archive (TCIA) database for 73 patients who underwent preoperative MRI procedures, and used the Least Absolute Shrinkage and Selection Operator (LASSO) to select the best imaging features for constructing an MRI-based radiomics model for evaluating immune cell infiltration levels in breast cancer patients. RESULTS According to the results of Cox regression analysis, M2 macrophages were identified as an independent prognostic factor for BRCA patients (HR = 32.288, 95% CI: 3.100-357.478). A total of nine significant features were selected to calculate the radiomics-based score. We established an intratumoral model with AUCs (95% CI) of 0.662 (0.495-0.802) and 0.678 (0.438-0.901) in the training and testing cohorts, respectively. Additionally, a peritumoral model was created with AUCs (95% CI) of 0.826 (0.710-0.924) and 0.752 (0.525-0.957), and a combined model was established with AUCs (95% CI) of 0.843 (0.723-0.938) and 0.744 (0.491-0.965). The peritumoral model demonstrated the highest diagnostic efficacy, with an accuracy, sensitivity, and specificity of 0.773, 0.727, and 0.818, respectively, in its testing cohort. CONCLUSION The MRI-based radiomics model has the potential to evaluate the degree of immune cell infiltration in breast cancer patients, offering a non-invasive imaging biomarker for assessing the tumor microenvironment in this disease.
Collapse
Affiliation(s)
- Hua Qian
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaojing Ren
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhen Fang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruixin Zhang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yangyang Bu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Changyu Zhou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), China , 54 Youdian Road, Hangzhou, 310006, Hangzhou, China.
- School of the First Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
24
|
Caloro E, Gnocchi G, Quarrella C, Ce M, Carrafiello G, Cellina M. Artificial Intelligence in Bone Metastasis Imaging: Recent Progresses from Diagnosis to Treatment - A Narrative Review. Crit Rev Oncog 2024; 29:77-90. [PMID: 38505883 DOI: 10.1615/critrevoncog.2023050470] [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: 03/21/2024]
Abstract
The introduction of artificial intelligence (AI) represents an actual revolution in the radiological field, including bone lesion imaging. Bone lesions are often detected both in healthy and oncological patients and the differential diagnosis can be challenging but decisive, because it affects the diagnostic and therapeutic process, especially in case of metastases. Several studies have already demonstrated how the integration of AI-based tools in the current clinical workflow could bring benefits to patients and to healthcare workers. AI technologies could help radiologists in early bone metastases detection, increasing the diagnostic accuracy and reducing the overdiagnosis and the number of unnecessary deeper investigations. In addition, radiomics and radiogenomics approaches could go beyond the qualitative features, visible to the human eyes, extrapolating cancer genomic and behavior information from imaging, in order to plan a targeted and personalized treatment. In this article, we want to provide a comprehensive summary of the most promising AI applications in bone metastasis imaging and their role from diagnosis to treatment and prognosis, including the analysis of future challenges and new perspectives.
Collapse
Affiliation(s)
- Elena Caloro
- Università degli studi di Milano, via Festa del Perdono, 7, 20122 Milan, Italy
| | - Giulia Gnocchi
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Cettina Quarrella
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Maurizio Ce
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, 20122 Milan, Italy
| | - Gianpaolo Carrafiello
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy; Radiology Department, Fondazione IRCCS Cà Granda, Policlinico di Milano Ospedale Maggiore, Università di Milano, 20122 Milan, Italy
| | - Michaela Cellina
- Radiology Department, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, Milano, Piazza Principessa Clotilde 3, 20121, Milan, Italy
| |
Collapse
|
25
|
Cellina M, De Padova G, Caldarelli N, Libri D, Cè M, Martinenghi C, Alì M, Papa S, Carrafiello G. Artificial Intelligence in Lung Cancer Imaging: From Data to Therapy. Crit Rev Oncog 2024; 29:1-13. [PMID: 38505877 DOI: 10.1615/critrevoncog.2023050439] [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: 03/21/2024]
Abstract
Lung cancer remains a global health challenge, leading to substantial morbidity and mortality. While prevention and early detection strategies have improved, the need for precise diagnosis, prognosis, and treatment remains crucial. In this comprehensive review article, we explore the role of artificial intelligence (AI) in reshaping the management of lung cancer. AI may have different potential applications in lung cancer characterization and outcome prediction. Manual segmentation is a time-consuming task, with high inter-observer variability, that can be replaced by AI-based approaches, including deep learning models such as U-Net, BCDU-Net, and others, to quantify lung nodules and cancers objectively and to extract radiomics features for the characterization of the tissue. AI models have also demonstrated their ability to predict treatment responses, such as immunotherapy and targeted therapy, by integrating radiomic features with clinical data. Additionally, AI-based prognostic models have been developed to identify patients at higher risk and personalize treatment strategies. In conclusion, this review article provides a comprehensive overview of the current state of AI applications in lung cancer management, spanning from segmentation and virtual biopsy to outcome prediction. The evolving role of AI in improving the precision and effectiveness of lung cancer diagnosis and treatment underscores its potential to significantly impact clinical practice and patient outcomes.
Collapse
Affiliation(s)
- Michaela Cellina
- Radiology Department, Fatebenefratelli Hospital, ASST Fatebenefratelli Sacco, Milano, Piazza Principessa Clotilde 3, 20121, Milan, Italy
| | - Giuseppe De Padova
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Nazarena Caldarelli
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Dario Libri
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Maurizio Cè
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy
| | - Carlo Martinenghi
- Radiology Department, Ospedale San Raffaele, Via Olgettina, 60 - 20132 Milan, Italy
| | - Marco Alì
- Radiology Unit, CDI, Centro Diagnostico Italiano, Via Simone Saint Bon, 20, 20147 Milan, Italy
| | - Sergio Papa
- Radiology Unit, CDI, Centro Diagnostico Italiano, Via Simone Saint Bon, 20, 20147 Milan, Italy
| | - Gianpaolo Carrafiello
- Postgraduation School in Radiodiagnostics, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122 Milan, Italy; Radiology Department, Fondazione IRCCS Cà Granda, Policlinico di Milano Ospedale Maggiore, Università di Milano, 20122 Milan, Italy
| |
Collapse
|
26
|
Ghimire P, Kinnersley B, Karami G, Arumugam P, Houlston R, Ashkan K, Modat M, Booth TC. Radiogenomic biomarkers for immunotherapy in glioblastoma: A systematic review of magnetic resonance imaging studies. Neurooncol Adv 2024; 6:vdae055. [PMID: 38680991 PMCID: PMC11046988 DOI: 10.1093/noajnl/vdae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024] Open
Abstract
Background Immunotherapy is an effective "precision medicine" treatment for several cancers. Imaging signatures of the underlying genome (radiogenomics) in glioblastoma patients may serve as preoperative biomarkers of the tumor-host immune apparatus. Validated biomarkers would have the potential to stratify patients during immunotherapy clinical trials, and if trials are beneficial, facilitate personalized neo-adjuvant treatment. The increased use of whole genome sequencing data, and the advances in bioinformatics and machine learning make such developments plausible. We performed a systematic review to determine the extent of development and validation of immune-related radiogenomic biomarkers for glioblastoma. Methods A systematic review was performed following PRISMA guidelines using the PubMed, Medline, and Embase databases. Qualitative analysis was performed by incorporating the QUADAS 2 tool and CLAIM checklist. PROSPERO registered: CRD42022340968. Extracted data were insufficiently homogenous to perform a meta-analysis. Results Nine studies, all retrospective, were included. Biomarkers extracted from magnetic resonance imaging volumes of interest included apparent diffusion coefficient values, relative cerebral blood volume values, and image-derived features. These biomarkers correlated with genomic markers from tumor cells or immune cells or with patient survival. The majority of studies had a high risk of bias and applicability concerns regarding the index test performed. Conclusions Radiogenomic immune biomarkers have the potential to provide early treatment options to patients with glioblastoma. Targeted immunotherapy, stratified by these biomarkers, has the potential to allow individualized neo-adjuvant precision treatment options in clinical trials. However, there are no prospective studies validating these biomarkers, and interpretation is limited due to study bias with little evidence of generalizability.
Collapse
Affiliation(s)
- Prajwal Ghimire
- Department of Neurosurgery, Kings College Hospital NHS Foundation Trust, London, UK
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Ben Kinnersley
- Department of Oncology, University College London, London, UK
| | | | | | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, UK
| | - Keyoumars Ashkan
- Department of Neurosurgery, Kings College Hospital NHS Foundation Trust, London, UK
| | - Marc Modat
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Thomas C Booth
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| |
Collapse
|
27
|
Zhong J, Kobus M, Maitre P, Datta A, Eccles C, Dubec M, McHugh D, Buckley D, Scarsbrook A, Hoskin P, Henry A, Choudhury A. MRI-guided Pelvic Radiation Therapy: A Primer for Radiologists. Radiographics 2023; 43:e230052. [PMID: 37796729 DOI: 10.1148/rg.230052] [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] [Indexed: 10/07/2023]
Abstract
Radiation therapy (RT) is a core pillar of oncologic treatment, and half of all patients with cancer receive this therapy as a curative or palliative treatment. The recent integration of MRI into the RT workflow has led to the advent of MRI-guided RT (MRIgRT). Using MRI rather than CT has clear advantages for guiding RT to pelvic tumors, including superior soft-tissue contrast, improved organ motion visualization, and the potential to image tumor phenotypic characteristics to identify the most aggressive or treatment-resistant areas, which can be targeted with a more focal higher radiation dose. Radiologists should be familiar with the potential uses of MRI in planning pelvic RT; the various RT techniques used, such as brachytherapy and external beam RT; and the impact of MRIgRT on treatment paradigms. Current clinical experience with and the evidence base for MRIgRT in the settings of prostate, cervical, and bladder cancer are discussed, and examples of treated cases are illustrated. In addition, the benefits of MRIgRT, such as real-time online adaptation of RT (during treatment) and interfraction and/or intrafraction adaptation to organ motion, as well as how MRIgRT can decrease toxic effects and improve oncologic outcomes, are highlighted. MRIgRT is particularly beneficial for treating mobile pelvic structures, and real-time adaptive RT for tumors can be achieved by using advanced MRI-guided linear accelerator systems to spare organs at risk. Future opportunities for development of biologically driven adapted RT with use of functional MRI sequences and radiogenomic approaches also are outlined. ©RSNA, 2023 Quiz questions for this article are available in the supplemental material.
Collapse
Affiliation(s)
- Jim Zhong
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| | - Marta Kobus
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| | - Priyamvada Maitre
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| | - Anubhav Datta
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| | - Cynthia Eccles
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| | - Michael Dubec
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| | - Damien McHugh
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| | - David Buckley
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| | - Andrew Scarsbrook
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| | - Peter Hoskin
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| | - Ann Henry
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| | - Ananya Choudhury
- From the Leeds Institute of Medical Research (J.Z., A.S., A.H.) and Department of Biomedical Imaging (D.B.), University of Leeds, 6 Clarendon Way, Woodhouse, Leeds LS2 9LH, England; Leeds Cancer Centre, St James's University Hospital, Leeds, England (J.Z., A.S., A.H.); Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany (M.K.); Radiation Therapy Research Group (M.K., P.M., A.D., C.E., M.D., P.H., A.C.) and Division of Cancer Sciences (D.M.), University of Manchester, Manchester, England; and The Christie NHS Foundation Trust, Manchester, England (P.M., C.E., M.D., D.M., P.H., A.C.)
| |
Collapse
|
28
|
Hussain S, Lafarga-Osuna Y, Ali M, Naseem U, Ahmed M, Tamez-Peña JG. Deep learning, radiomics and radiogenomics applications in the digital breast tomosynthesis: a systematic review. BMC Bioinformatics 2023; 24:401. [PMID: 37884877 PMCID: PMC10605943 DOI: 10.1186/s12859-023-05515-6] [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: 04/07/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Recent advancements in computing power and state-of-the-art algorithms have helped in more accessible and accurate diagnosis of numerous diseases. In addition, the development of de novo areas in imaging science, such as radiomics and radiogenomics, have been adding more to personalize healthcare to stratify patients better. These techniques associate imaging phenotypes with the related disease genes. Various imaging modalities have been used for years to diagnose breast cancer. Nonetheless, digital breast tomosynthesis (DBT), a state-of-the-art technique, has produced promising results comparatively. DBT, a 3D mammography, is replacing conventional 2D mammography rapidly. This technological advancement is key to AI algorithms for accurately interpreting medical images. OBJECTIVE AND METHODS This paper presents a comprehensive review of deep learning (DL), radiomics and radiogenomics in breast image analysis. This review focuses on DBT, its extracted synthetic mammography (SM), and full-field digital mammography (FFDM). Furthermore, this survey provides systematic knowledge about DL, radiomics, and radiogenomics for beginners and advanced-level researchers. RESULTS A total of 500 articles were identified, with 30 studies included as the set criteria. Parallel benchmarking of radiomics, radiogenomics, and DL models applied to the DBT images could allow clinicians and researchers alike to have greater awareness as they consider clinical deployment or development of new models. This review provides a comprehensive guide to understanding the current state of early breast cancer detection using DBT images. CONCLUSION Using this survey, investigators with various backgrounds can easily seek interdisciplinary science and new DL, radiomics, and radiogenomics directions towards DBT.
Collapse
Affiliation(s)
- Sadam Hussain
- School of Engineering and Sciences, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, 64849, Monterrey, Mexico.
| | - Yareth Lafarga-Osuna
- School of Engineering and Sciences, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, 64849, Monterrey, Mexico
| | - Mansoor Ali
- School of Engineering and Sciences, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, 64849, Monterrey, Mexico
| | - Usman Naseem
- College of Science and Engineering, James Cook University, Cairns, Australia
| | - Masroor Ahmed
- School of Engineering and Sciences, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, 64849, Monterrey, Mexico
| | - Jose Gerardo Tamez-Peña
- School of Medicine and Health Sciences, Tecnológico de Monterrey, Ave. Eugenio Garza Sada 2501, 64849, Monterrey, Mexico
| |
Collapse
|
29
|
Liu Z, Duan T, Zhang Y, Weng S, Xu H, Ren Y, Zhang Z, Han X. Radiogenomics: a key component of precision cancer medicine. Br J Cancer 2023; 129:741-753. [PMID: 37414827 PMCID: PMC10449908 DOI: 10.1038/s41416-023-02317-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 05/02/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Radiogenomics, focusing on the relationship between genomics and imaging phenotypes, has been widely applied to address tumour heterogeneity and predict immune responsiveness and progression. It is an inevitable consequence of current trends in precision medicine, as radiogenomics costs less than traditional genetic sequencing and provides access to whole-tumour information rather than limited biopsy specimens. By providing voxel-by-voxel genetic information, radiogenomics can allow tailored therapy targeting a complete, heterogeneous tumour or set of tumours. In addition to quantifying lesion characteristics, radiogenomics can also be used to distinguish benign from malignant entities, as well as patient characteristics, to better stratify patients according to disease risk, thereby enabling more precise imaging and screening. Here, we have characterised the radiogenomic application in precision medicine using a multi-omic approach. we outline the main applications of radiogenomics in diagnosis, treatment planning and evaluations in the field of oncology with the aim of developing quantitative and personalised medicine. Finally, we discuss the challenges in the field of radiogenomics and the scope and clinical applicability of these methods.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, 450052, Zhengzhou, Henan, China
| | - Tian Duan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
30
|
Kersch CN, Muldoon LL, Claunch CJ, Fu R, Schwartz D, Cha S, Starkey J, Neuwelt EA, Barajas RF. Multiparametric magnetic resonance imaging discerns glioblastoma immune microenvironmental heterogeneity. Neuroradiol J 2023:19714009231163560. [PMID: 37306690 DOI: 10.1177/19714009231163560] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023] Open
Abstract
RATIONALE AND OBJECTIVE Poor clinical outcomes for patients with glioblastoma are in part due to dysfunction of the tumor-immune microenvironment. An imaging approach able to characterize immune microenvironmental signatures could provide a framework for biologically based patient stratification and response assessment. We hypothesized spatially distinct gene expression networks can be distinguished by multiparametric Magnetic Resonance Imaging (MRI) phenotypes. MATERIALS AND METHODS Patients with newly diagnosed glioblastoma underwent image-guided tissue sampling allowing for co-registration of MRI metrics with gene expression profiles. MRI phenotypes based on gadolinium contrast enhancing lesion (CEL) and non-enhancing lesion (NCEL) regions were subdivided based on imaging parameters (relative cerebral blood volume (rCBV) and apparent diffusion coefficient (ADC)). Gene set enrichment analysis and immune cell type abundance was estimated using CIBERSORT methodology. Significance thresholds were set at a p-value cutoff 0.005 and an FDR q-value cutoff of 0.1. RESULTS Thirteen patients (eight men, five women, mean age 58 ± 11 years) provided 30 tissue samples (16 CEL and 14 NCEL). Six non-neoplastic gliosis samples differentiated astrocyte repair from tumor associated gene expression. MRI phenotypes displayed extensive transcriptional variance reflecting biological networks, including multiple immune pathways. CEL regions demonstrated higher immunologic signature expression than NCEL, while NCEL regions demonstrated stronger immune signature expression levels than gliotic non-tumor brain. Incorporation of rCBV and ADC metrics identified sample clusters with differing immune microenvironmental signatures. CONCLUSION Taken together, our study demonstrates that MRI phenotypes provide an approach for non-invasively characterizing tumoral and immune microenvironmental glioblastoma gene expression networks.
Collapse
Affiliation(s)
- Cymon N Kersch
- Department of Neurology, Blood-Brain Barrier Program, Oregon Health & Sciences University, USA
- Department of Radiation Medicine, Oregon Health & Sciences University, USA
| | - Leslie L Muldoon
- Department of Neurology, Blood-Brain Barrier Program, Oregon Health & Sciences University, USA
| | - Cheryl J Claunch
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health & Sciences University, USA
| | - Rongwei Fu
- OHSU-PSU School of Public Health, Oregon Health & Sciences University, USA
| | - Daniel Schwartz
- Advanced Imaging Research Center, Oregon Health & Sciences University, USA
- Department of Neurology, Layton Aging and Alzheimer's Disease Center, Oregon Health & Sciences University, USA
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, University of California San Francisco, USA
| | - Jay Starkey
- Department of Radiology, Oregon Health & Sciences University, USA
| | - Edward A Neuwelt
- Department of Neurology, Blood-Brain Barrier Program, Oregon Health & Sciences University, USA
- Department of Neurosurgery, Oregon Health & Sciences University, USA
- Office of Research and Development, Department of Veterans Affairs Medical Center, USA
| | - Ramon F Barajas
- Advanced Imaging Research Center, Oregon Health & Sciences University, USA
- Department of Radiology, Oregon Health & Sciences University, USA
- Knight Cancer Institute, Oregon Health & Sciences University, USA
| |
Collapse
|
31
|
Demirel E, Dilek O. Relationship between body composition and PBRM1 mutations in clear cell renal cell carcinoma: a propensity score matching analysis. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20220415. [PMID: 37222312 DOI: 10.1590/1806-9282.20220415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/20/2023] [Indexed: 05/25/2023]
Abstract
OBJECTIVE This study aimed to examine the relationship between body muscle and adipose tissue composition in clear cell renal cell carcinoma patients with PBRM1 gene mutation. METHODS Cancer Genome Atlas Kidney clear cell renal cell carcinoma and Clinical Proteomic Tumor Analysis Consortium clear cell renal cell carcinoma collections were retrieved from the Cancer Imaging Archive. A total of 291 clear cell renal cell carcinoma patients were included in the study retrospectively. Patients' characteristics were obtained from Cancer Imaging Archive. Body composition was assessed with abdominal computed tomography using the automated artificial intelligence software (AID-U™, iAID Inc., Seoul, Korea). Body composition parameters of the patients were calculated. To investigate the net effect of body composition, the propensity score matching procedure was applied over age, gender, and T-stage parameters. RESULTS Of the patients, 184 were males and 107 were females. Mutations in the PBRM1 gene were detected in 77 of the patients. While there was no difference in adipose tissue areas between the PBRM1 mutation group and those without PBRM1 mutation, statistically significant differences were found in normal attenuated muscle area parameters. CONCLUSION This study shows that there was no difference between adipose tissue areas in patients with PBMR1 mutation, but normal attenuated muscle area was found to be higher in PBRM1 patients.
Collapse
Affiliation(s)
- Emin Demirel
- Emirdag City of Hospital, Department of Radiology - Afyonkarahisar, Turkey
| | - Okan Dilek
- University of Health Sciences, Adana City Training and Research Hospital, Department of Radiology - Adana, Turkey
| |
Collapse
|
32
|
Guan X, Lu N, Zhang J. Computed Tomography-Based Deep Learning Nomogram Can Accurately Predict Lymph Node Metastasis in Gastric Cancer. Dig Dis Sci 2023; 68:1473-1481. [PMID: 35909203 PMCID: PMC10102043 DOI: 10.1007/s10620-022-07640-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/18/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND Computed tomography is the most commonly used imaging modality for preoperative assessment of lymph node status, but the reported accuracy is unsatisfactory. AIMS To evaluate and verify the predictive performance of computed tomography deep learning on the presurgical evaluation of lymph node metastasis in patients with gastric cancer. METHODS 347 patients were retrospectively selected (training cohort: 242, test cohort: 105). The enhanced computed tomography arterial phase images of gastric cancer were used for lesion segmentation, radiomics and deep learning feature extraction. Three methods were used for feature selection. Support vector machine (SVM) or random forest (RF) was used to build models. The classification performance of the models was evaluated using the area under the receiver operating characteristic curve (AUC). We also established a nomogram that included clinical predictors. RESULTS The model based on ResNet50-RF showed favorable classification performance and was verified in the test cohort (AUC = 0.9803). The nomogram based on deep learning feature scores and the lymph node status reported by computed tomography showed excellent discrimination. AUC of 0.9978 was achieved in the training cohort and verified in the test cohort (AUC = 0.9914). Decision analysis curve showed the value of nomogram in clinical application. CONCLUSION The computed tomography-based deep learning nomogram can accurately and effectively evaluate lymph node metastasis in patients with gastric cancer before surgery.
Collapse
Affiliation(s)
- Xiao Guan
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No. 121, Jiangjiayuan Road, Nanjing, 210011 Jiangsu China
| | - Na Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No. 121, Jiangjiayuan Road, Nanjing, 210011 Jiangsu China
| | - Jianping Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No. 121, Jiangjiayuan Road, Nanjing, 210011 Jiangsu China
| |
Collapse
|
33
|
Panico C, Avesani G, Zormpas-Petridis K, Rundo L, Nero C, Sala E. Radiomics and Radiogenomics of Ovarian Cancer. Radiol Clin North Am 2023; 61:749-760. [PMID: 37169435 DOI: 10.1016/j.rcl.2023.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Ovarian cancer, one of the deadliest gynecologic malignancies, is characterized by high intra- and inter-site genomic and phenotypic heterogeneity. The traditional information provided by the conventional interpretation of diagnostic imaging studies cannot adequately represent this heterogeneity. Radiomics analyses can capture the complex patterns related to the microstructure of the tissues and provide quantitative information about them. This review outlines how radiomics and its integration with other quantitative biological information, like genomics and proteomics, can impact the clinical management of ovarian cancer.
Collapse
|
34
|
Nabavizadeh A, Barkovich MJ, Mian A, Ngo V, Kazerooni AF, Villanueva-Meyer JE. Current state of pediatric neuro-oncology imaging, challenges and future directions. Neoplasia 2023; 37:100886. [PMID: 36774835 PMCID: PMC9945752 DOI: 10.1016/j.neo.2023.100886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/20/2023] [Accepted: 02/05/2023] [Indexed: 02/12/2023]
Abstract
Imaging plays a central role in neuro-oncology including primary diagnosis, treatment planning, and surveillance of tumors. The emergence of quantitative imaging and radiomics provided an uprecedented opportunity to compile mineable databases that can be utilized in a variety of applications. In this review, we aim to summarize the current state of conventional and advanced imaging techniques, standardization efforts, fast protocols, contrast and sedation in pediatric neuro-oncologic imaging, radiomics-radiogenomics, multi-omics and molecular imaging approaches. We will also address the existing challenges and discuss future directions.
Collapse
Affiliation(s)
- Ali Nabavizadeh
- Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
| | - Matthew J Barkovich
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Ali Mian
- Division of Neuroradiology, Mallinckrodt Institute of Radiology, Washington University in St. Louis, Missouri, USA
| | - Van Ngo
- Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
35
|
Ferro M, Musi G, Marchioni M, Maggi M, Veccia A, Del Giudice F, Barone B, Crocetto F, Lasorsa F, Antonelli A, Schips L, Autorino R, Busetto GM, Terracciano D, Lucarelli G, Tataru OS. Radiogenomics in Renal Cancer Management-Current Evidence and Future Prospects. Int J Mol Sci 2023; 24:4615. [PMID: 36902045 PMCID: PMC10003020 DOI: 10.3390/ijms24054615] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Renal cancer management is challenging from diagnosis to treatment and follow-up. In cases of small renal masses and cystic lesions the differential diagnosis of benign or malignant tissues has potential pitfalls when imaging or even renal biopsy is applied. The recent artificial intelligence, imaging techniques, and genomics advancements have the ability to help clinicians set the stratification risk, treatment selection, follow-up strategy, and prognosis of the disease. The combination of radiomics features and genomics data has achieved good results but is currently limited by the retrospective design and the small number of patients included in clinical trials. The road ahead for radiogenomics is open to new, well-designed prospective studies, with large cohorts of patients required to validate previously obtained results and enter clinical practice.
Collapse
Affiliation(s)
- Matteo Ferro
- Department of Urology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| | - Gennaro Musi
- Department of Urology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| | - Michele Marchioni
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio, University of Chieti, 66100 Chieti, Italy
- Urology Unit, SS. Annunziata Hospital, 66100 Chieti, Italy
- Department of Urology, ASL Abruzzo 2, 66100 Chieti, Italy
| | - Martina Maggi
- Department of Maternal Infant and Urologic Sciences, Policlinico Umberto I Hospital, University of Rome, 00161 Rome, Italy
| | - Alessandro Veccia
- Department of Urology, Azienda Ospedaliera Universitaria Integrata of Verona, University of Verona, 37126 Verona, Italy
| | - Francesco Del Giudice
- Department of Maternal Infant and Urologic Sciences, Policlinico Umberto I Hospital, University of Rome, 00161 Rome, Italy
| | - Biagio Barone
- Department of Neurosciences and Reproductive Sciences and Odontostomatology, University of Naples Federico II, 80131 Naples, Italy
| | - Felice Crocetto
- Department of Neurosciences and Reproductive Sciences and Odontostomatology, University of Naples Federico II, 80131 Naples, Italy
| | - Francesco Lasorsa
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Alessandro Antonelli
- Department of Urology, Azienda Ospedaliera Universitaria Integrata of Verona, University of Verona, 37126 Verona, Italy
| | - Luigi Schips
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio, University of Chieti, 66100 Chieti, Italy
- Urology Unit, SS. Annunziata Hospital, 66100 Chieti, Italy
- Department of Urology, ASL Abruzzo 2, 66100 Chieti, Italy
| | | | - Gian Maria Busetto
- Department of Urology and Renal Transplantation, University of Foggia, 71122 Foggia, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Octavian Sabin Tataru
- Department of Simulation Applied in Medicine, The Institution Organizing University Doctoral Studies (I.O.S.U.D.), George Emil Palade University of Medicine, Pharmacy, Sciences, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania
| |
Collapse
|
36
|
Qureshi SA, Hussain L, Ibrar U, Alabdulkreem E, Nour MK, Alqahtani MS, Nafie FM, Mohamed A, Mohammed GP, Duong TQ. Radiogenomic classification for MGMT promoter methylation status using multi-omics fused feature space for least invasive diagnosis through mpMRI scans. Sci Rep 2023; 13:3291. [PMID: 36841898 PMCID: PMC9961309 DOI: 10.1038/s41598-023-30309-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/21/2023] [Indexed: 02/27/2023] Open
Abstract
Accurate radiogenomic classification of brain tumors is important to improve the standard of diagnosis, prognosis, and treatment planning for patients with glioblastoma. In this study, we propose a novel two-stage MGMT Promoter Methylation Prediction (MGMT-PMP) system that extracts latent features fused with radiomic features predicting the genetic subtype of glioblastoma. A novel fine-tuned deep learning architecture, namely Deep Learning Radiomic Feature Extraction (DLRFE) module, is proposed for latent feature extraction that fuses the quantitative knowledge to the spatial distribution and the size of tumorous structure through radiomic features: (GLCM, HOG, and LBP). The application of the novice rejection algorithm has been found significantly effective in selecting and isolating the negative training instances out of the original dataset. The fused feature vectors are then used for training and testing by k-NN and SVM classifiers. The 2021 RSNA Brain Tumor challenge dataset (BraTS-2021) consists of four structural mpMRIs, viz. fluid-attenuated inversion-recovery, T1-weighted, T1-weighted contrast enhancement, and T2-weighted. We evaluated the classification performance, for the very first time in published form, in terms of measures like accuracy, F1-score, and Matthews correlation coefficient. The Jackknife tenfold cross-validation was used for training and testing BraTS-2021 dataset validation. The highest classification performance is (96.84 ± 0.09)%, (96.08 ± 0.10)%, and (97.44 ± 0.14)% as accuracy, sensitivity, and specificity respectively to detect MGMT methylation status for patients suffering from glioblastoma. Deep learning feature extraction with radiogenomic features, fusing imaging phenotypes and molecular structure, using rejection algorithm has been found to perform outclass capable of detecting MGMT methylation status of glioblastoma patients. The approach relates the genomic variation with radiomic features forming a bridge between two areas of research that may prove useful for clinical treatment planning leading to better outcomes.
Collapse
Affiliation(s)
- Shahzad Ahmad Qureshi
- Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Islamabad, Pakistan.
| | - Lal Hussain
- Department of Computer Science and IT, Neelum Campus, The University of Azad Jammu and Kashmir, Muzaffarabad, Azad Kashmir, Pakistan. .,Department of Computer Science and IT, King Abdullah Campus, The University of Azad Jammu and Kashmir, Muzaffarabad, Azad Kashmir, Pakistan. .,Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 111 East 210th Street, Bronx, NY, 10467, USA.
| | - Usama Ibrar
- grid.461150.7Farooq Hospital, Lahore, Pakistan
| | - Eatedal Alabdulkreem
- grid.449346.80000 0004 0501 7602Department of Computer Sciences, College of Computer and Information Sciences, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671 Saudi Arabia
| | - Mohamed K. Nour
- grid.412832.e0000 0000 9137 6644Department of Computer Sciences, College of Computing and Information System, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Mohammed S. Alqahtani
- grid.412144.60000 0004 1790 7100Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, 61421 Saudi Arabia
| | - Faisal Mohammed Nafie
- grid.449051.d0000 0004 0441 5633Department of Computer Science, College of Science and Humanities at Alghat, Majmaah University, Al-Majmaah, 11952 Saudi Arabia
| | - Abdullah Mohamed
- grid.440865.b0000 0004 0377 3762Research Centre, Future University in Egypt, New Cairo, 11845 Egypt
| | - Gouse Pasha Mohammed
- grid.449553.a0000 0004 0441 5588Department of Computer and Self Development, Preparatory Year Deanship, Prince Sattam Bin Abdulaziz University, AlKharj, Saudi Arabia
| | - Tim Q. Duong
- grid.240283.f0000 0001 2152 0791Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467 USA
| |
Collapse
|
37
|
Yan J, Sun Q, Tan X, Liang C, Bai H, Duan W, Mu T, Guo Y, Qiu Y, Wang W, Yao Q, Pei D, Zhao Y, Liu D, Duan J, Chen S, Sun C, Wang W, Liu Z, Hong X, Wang X, Guo Y, Xu Y, Liu X, Cheng J, Li ZC, Zhang Z. Image-based deep learning identifies glioblastoma risk groups with genomic and transcriptomic heterogeneity: a multi-center study. Eur Radiol 2023; 33:904-914. [PMID: 36001125 DOI: 10.1007/s00330-022-09066-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVES To develop and validate a deep learning imaging signature (DLIS) for risk stratification in patients with multiforme (GBM), and to investigate the biological pathways and genetic alterations underlying the DLIS. METHODS The DLIS was developed from multi-parametric MRI based on a training set (n = 600) and validated on an internal validation set (n = 164), an external test set 1 (n = 100), an external test set 2 (n = 161), and a public TCIA set (n = 88). A co-profiling framework based on a radiogenomics analysis dataset (n = 127) using multiscale high-dimensional data, including imaging, transcriptome, and genome, was established to uncover the biological pathways and genetic alterations underpinning the DLIS. RESULTS The DLIS was associated with survival (log-rank p < 0.001) and was an independent predictor (p < 0.001). The integrated nomogram incorporating the DLIS achieved improved C indices than the clinicomolecular nomogram (net reclassification improvement 0.39, p < 0.001). DLIS significantly correlated with core pathways of GBM (apoptosis and cell cycle-related P53 and RB pathways, and cell proliferation-related RTK pathway), as well as key genetic alterations (del_CDNK2A). The prognostic value of DLIS-correlated genes was externally confirmed on TCGA/CGGA sets (p < 0.01). CONCLUSIONS Our study offers a biologically interpretable deep learning predictor of survival outcomes in patients with GBM, which is crucial for better understanding GBM patient's prognosis and guiding individualized treatment. KEY POINTS • MRI-based deep learning imaging signature (DLIS) stratifies GBM into risk groups with distinct molecular characteristics. • DLIS is associated with P53, RB, and RTK pathways and del_CDNK2A mutation. • The prognostic value of DLIS-correlated pathway genes is externally demonstrated.
Collapse
Affiliation(s)
- Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Qiuchang Sun
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiangliang Tan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaofeng Liang
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Hongmin Bai
- Department of Neurosurgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, China
| | - Wenchao Duan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Tianhao Mu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.,HaploX Biotechnology, Shenzhen, Guangdong, China
| | - Yang Guo
- Department of Neurosurgery, Henan Provincial Hospital, Zhengzhou, 450052, Henan Province, China
| | - Yuning Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan Province, China
| | - Qiaoli Yao
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Dongling Pei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Yuanshen Zhao
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Danni Liu
- HaploX Biotechnology, Shenzhen, Guangdong, China
| | - Jingxian Duan
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Shifu Chen
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.,HaploX Biotechnology, Shenzhen, Guangdong, China
| | - Chen Sun
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Wenqing Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Zhen Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Xuanke Hong
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Xiangxiang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Yu Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China.
| | - Zhi-Cheng Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Shenzhen United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, 518045, China.
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China.
| |
Collapse
|
38
|
He XM, Zhao JX, He DL, Ren JL, Zhao LP, Huang G. Radiogenomics study to predict the nuclear grade of renal clear cell carcinoma. Eur J Radiol Open 2023; 10:100476. [PMID: 36793772 PMCID: PMC9922923 DOI: 10.1016/j.ejro.2023.100476] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 01/30/2023] Open
Abstract
Purpose To develop models based on radiomics and genomics for predicting the histopathologic nuclear grade with localized clear cell renal cell carcinoma (ccRCC) and to assess whether macro-radiomics models can predict the microscopic pathological changes. Method In this multi-institutional retrospective study, a computerized tomography (CT) radiomic model for nuclear grade prediction was developed. Utilizing a genomics analysis cohort, nuclear grade-associated gene modules were identified, and a gene model was constructed based on top 30 hub mRNA to predict the nuclear grade. Using a radiogenomic development cohort, biological pathways were enriched by hub genes and a radiogenomic map was created. Results The four-features-based SVM model predicted nuclear grade with an area under the curve (AUC) score of 0.94 in validation sets, while a five-gene-based model predicted nuclear grade with an AUC of 0.73 in the genomics analysis cohort. A total of five gene modules were identified to be associated with the nuclear grade. Radiomic features were only associated with 271 out of 603 genes in five gene modules and eight top 30 hub genes. Differences existed in the enrichment pathway between associated and un-associated with radiomic features, which were associated with two genes of five-gene signatures in the mRNA model. Conclusion The CT radiomics models exhibited higher predictive performance than mRNA models. The association between radiomic features and mRNA related to nuclear grade is not universal.
Collapse
Key Words
- Computer Applications
- FDR, False discovery rate
- GLRLM, Gray level run length matrix
- GLSZM, Gray level size matrix
- KEGG, KOBAS-Kyoto Encyclopedia of Genes and Genomes
- Kidney
- NGTDM, Neighborhood gray tone difference matrix
- Neoplasms-Primary
- PPI, Protein-Protein Interaction Networks
- Pathological nuclear grade
- Radiogenomics
- Radiomics
- TCGA, The cancer genome atlas
- TCIA, The cancer imaging archive
- WGCNA, Weighted gene co-expression network
- WHO/ISUP, World Health Organization and International Society of Urological Pathology
- ccRCC, Clear cell renal cell carcinoma
Collapse
Affiliation(s)
- Xuan-ming He
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jian-xin Zhao
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Di-liang He
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | | | - Lian-ping Zhao
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, China
| | - Gang Huang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, China,Corresponding author.
| |
Collapse
|
39
|
Kim JY, Jelinek J, Lee YH, Kim DH, Kang K, Ryu SH, Moon HR, Cho K, Rha SH, Cha JK, Issa JPJ, Kim J. Hypomethylation in MTNR1B: a novel epigenetic marker for atherosclerosis profiling using stenosis radiophenotype and blood inflammatory cells. Clin Epigenetics 2023; 15:11. [PMID: 36658621 PMCID: PMC9854223 DOI: 10.1186/s13148-023-01423-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/06/2023] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Changes in gene-specific promoter methylation may result from aging and environmental influences. Atherosclerosis is associated with aging and environmental effects. Thus, promoter methylation profiling may be used as an epigenetic tool to evaluate the impact of aging and the environment on atherosclerosis development. However, gene-specific methylation changes are currently inadequate epigenetic markers for predicting atherosclerosis and cardiovascular disease pathogenesis. RESULTS We profiled and validated changes in gene-specific promoter methylation associated with atherosclerosis using stenosis radiophenotypes of cranial vessels and blood inflammatory cells rather than direct sampling of atherosclerotic plaques. First, we profiled gene-specific promoter methylation changes using digital restriction enzyme analysis of methylation (DREAM) sequencing in peripheral blood mononuclear cells from eight samples each of cranial vessels with and without severe-stenosis radiophenotypes. Using DREAM sequencing profiling, 11 tags were detected in the promoter regions of the ACVR1C, ADCK5, EFNA2, ENOSF1, GLS2, KNDC1, MTNR1B, PACSIN3, PAX8-AS1, TLDC1, and ZNF7 genes. Using methylation evaluation, we found that EFNA2, ENOSF1, GLS2, KNDC1, MTNR1B, PAX8-AS1, and TLDC1 showed > 5% promoter methylation in non-plaque intima, atherosclerotic vascular tissues, and buffy coats. Using logistic regression analysis, we identified hypomethylation of MTNR1B as an independent variable for the stenosis radiophenotype prediction model by combining it with traditional atherosclerosis risk factors including age, hypertension history, and increases in creatinine, lipoprotein (a), and homocysteine. We performed fivefold cross-validation of the prediction model using 384 patients with ischemic stroke (50 [13%] no-stenosis and 334 [87%] > 1 stenosis radiophenotype). For the cross-validation, the training dataset included 70% of the dataset. The prediction model showed an accuracy of 0.887, specificity to predict stenosis radiophenotype of 0.940, sensitivity to predict no-stenosis radiophenotype of 0.533, and area under receiver operating characteristic curve of 0.877 to predict stenosis radiophenotype from the test dataset including 30% of the dataset. CONCLUSIONS We identified and validated MTNR1B hypomethylation as an epigenetic marker to predict cranial vessel atherosclerosis using stenosis radiophenotypes and blood inflammatory cells rather than direct atherosclerotic plaque sampling.
Collapse
Affiliation(s)
- Jee Yeon Kim
- grid.254230.20000 0001 0722 6377Department of Neurology and Neuroepigenetics Laboratory, College of Medicine and Hospital, Chungnam National University, 282 Moonhwaro, Joongku, Daejeon, 35015 South Korea
| | - Jaroslav Jelinek
- grid.282012.b0000 0004 0627 5048Coriell Institute for Medical Research, Camden, NJ USA
| | - Young Ho Lee
- grid.254230.20000 0001 0722 6377Department of Anatomy, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Dae Hyun Kim
- grid.412048.b0000 0004 0647 1081Department of Neurology, Dong-A University Hospital, Busan, South Korea
| | - Keunsoo Kang
- grid.411982.70000 0001 0705 4288Department of Microbiology, College of Science and Technology, Dankook University, Cheonan, South Korea
| | - Su Hyun Ryu
- grid.254230.20000 0001 0722 6377Department of Neurology and Neuroepigenetics Laboratory, College of Medicine and Hospital, Chungnam National University, 282 Moonhwaro, Joongku, Daejeon, 35015 South Korea
| | - Hye Rin Moon
- grid.254230.20000 0001 0722 6377Department of Neurology and Neuroepigenetics Laboratory, College of Medicine and Hospital, Chungnam National University, 282 Moonhwaro, Joongku, Daejeon, 35015 South Korea
| | - Kwangjo Cho
- grid.412048.b0000 0004 0647 1081Department of Thoracic and Cardiovascular Surgery, Dong-A University Hospital, Busan, South Korea
| | - Seo Hee Rha
- grid.412048.b0000 0004 0647 1081Department of Pathology, Dong-A University Hospital, Busan, South Korea
| | - Jae Kwan Cha
- grid.254230.20000 0001 0722 6377Department of Anatomy, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jean-Pierre J. Issa
- grid.282012.b0000 0004 0627 5048Coriell Institute for Medical Research, Camden, NJ USA
| | - Jei Kim
- grid.254230.20000 0001 0722 6377Department of Neurology and Neuroepigenetics Laboratory, College of Medicine and Hospital, Chungnam National University, 282 Moonhwaro, Joongku, Daejeon, 35015 South Korea ,grid.411665.10000 0004 0647 2279Daejeon-Chungnam Regional Cerebrovascular Center, Chungnam National University Hospital, Daejeon, South Korea
| |
Collapse
|
40
|
Stoller J, Kersch CN, Muldoon LL, Ambady P, Harrington CA, Fu R, Raslan AM, Dogan A, Neuwelt EA, Barajas RF. Deciphering spatially distinct immune microenvironments in glioblastoma using ferumoxytol and gadolinium-enhanced and FLAIR hyperintense MRI phenotypes. Neurooncol Adv 2023; 5:vdad148. [PMID: 38077209 PMCID: PMC10699850 DOI: 10.1093/noajnl/vdad148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024] Open
Abstract
Background MRI with gadolinium (Gd)-contrast agents is used to assess glioblastoma treatment response but does not specifically reveal heterogeneous biology or immune microenvironmental composition. Ferumoxytol (Fe) contrast is an iron nanoparticle that localizes glioblastoma macrophages and microglia. Therefore, we hypothesized that the use of Fe contrast improves upon standard Gd-based T1-weighted and T2/FLAIR analysis by specifically delineating immune processes. Methods In this, HIPAA-compliant institutional review board-approved prospective study, stereotactic biopsy samples were acquired from patients with treatment-naïve and recurrent glioblastoma based on MR imaging phenotypes; Gd and Fe T1 enhancement (Gd+, Fe+) or not (Gd-, Fe-), as well as T2-Flair hyperintensity (FLAIR+, FLAIR-). Analysis of genetic expression was performed with RNA microarrays. Imaging and genomic expression patterns were compared using false discovery rate statistics. Results MR imaging phenotypes defined a variety of immune pathways and Hallmark gene sets. Gene set enrichment analysis demonstrated that Gd+, Fe+, and FLAIR+ features were individually correlated with the same 7 immune process gene sets. Fe+ tissue showed the greatest degree of immune Hallmark gene sets compared to Gd+ or Flair+ tissues and had statistically elevated M2 polarized macrophages, among others. Importantly, the FLAIR+ Gd+ and Fe- imaging phenotypes did not demonstrate expression of immune Hallmark gene sets. Conclusions Our study demonstrates the potential of Fe and Gd-enhanced MRI phenotypes to reveal spatially distinct immune processes within glioblastoma. Fe improves upon the standard of care Gd enhancement by specifically localizing glioblastoma-associated inflammatory processes, providing valuable insights into tumor biology.
Collapse
Affiliation(s)
- Jared Stoller
- Department of Diagnostic Radiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Cymon N Kersch
- Department of Radiation Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Leslie L Muldoon
- Department of Neurology, Oregon Health and Science University, Portland, Oregon, USA
| | - Prakash Ambady
- Department of Oncology, Providence Hospital, Portland, Oregon, USA
| | - Christina A Harrington
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Rongwei Fu
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, Oregon, USA
| | - Ahmed M Raslan
- Department of Neurosurgery, Oregon Health and Science University, Portland, Oregon, USA
| | - Aclan Dogan
- Department of Neurosurgery, Oregon Health and Science University, Portland, Oregon, USA
| | - Edward A Neuwelt
- Department of Neurology, Oregon Health and Science University, Portland, Oregon, USA
- Department of Neurosurgery, Oregon Health and Science University, Portland, Oregon, USA
- Department of Veterans Affairs Medical Center, Portland, Oregon, USA
| | - Ramon F Barajas
- Department of Diagnostic Radiology, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
41
|
Sminia P, Guipaud O, Viktorsson K, Ahire V, Baatout S, Boterberg T, Cizkova J, Dostál M, Fernandez-Palomo C, Filipova A, François A, Geiger M, Hunter A, Jassim H, Edin NFJ, Jordan K, Koniarová I, Selvaraj VK, Meade AD, Milliat F, Montoro A, Politis C, Savu D, Sémont A, Tichy A, Válek V, Vogin G. Clinical Radiobiology for Radiation Oncology. RADIOBIOLOGY TEXTBOOK 2023:237-309. [DOI: 10.1007/978-3-031-18810-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
AbstractThis chapter is focused on radiobiological aspects at the molecular, cellular, and tissue level which are relevant for the clinical use of ionizing radiation (IR) in cancer therapy. For radiation oncology, it is critical to find a balance, i.e., the therapeutic window, between the probability of tumor control and the probability of side effects caused by radiation injury to the healthy tissues and organs. An overview is given about modern precision radiotherapy (RT) techniques, which allow optimal sparing of healthy tissues. Biological factors determining the width of the therapeutic window are explained. The role of the six typical radiobiological phenomena determining the response of both malignant and normal tissues in the clinic, the 6R’s, which are Reoxygenation, Redistribution, Repopulation, Repair, Radiosensitivity, and Reactivation of the immune system, is discussed. Information is provided on tumor characteristics, for example, tumor type, growth kinetics, hypoxia, aberrant molecular signaling pathways, cancer stem cells and their impact on the response to RT. The role of the tumor microenvironment and microbiota is described and the effects of radiation on the immune system including the abscopal effect phenomenon are outlined. A summary is given on tumor diagnosis, response prediction via biomarkers, genetics, and radiomics, and ways to selectively enhance the RT response in tumors. Furthermore, we describe acute and late normal tissue reactions following exposure to radiation: cellular aspects, tissue kinetics, latency periods, permanent or transient injury, and histopathology. Details are also given on the differential effect on tumor and late responding healthy tissues following fractionated and low dose rate irradiation as well as the effect of whole-body exposure.
Collapse
|
42
|
Ferro M, Crocetto F, Barone B, del Giudice F, Maggi M, Lucarelli G, Busetto GM, Autorino R, Marchioni M, Cantiello F, Crocerossa F, Luzzago S, Piccinelli M, Mistretta FA, Tozzi M, Schips L, Falagario UG, Veccia A, Vartolomei MD, Musi G, de Cobelli O, Montanari E, Tătaru OS. Artificial intelligence and radiomics in evaluation of kidney lesions: a comprehensive literature review. Ther Adv Urol 2023; 15:17562872231164803. [PMID: 37113657 PMCID: PMC10126666 DOI: 10.1177/17562872231164803] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/04/2023] [Indexed: 04/29/2023] Open
Abstract
Radiomics and artificial intelligence (AI) may increase the differentiation of benign from malignant kidney lesions, differentiation of angiomyolipoma (AML) from renal cell carcinoma (RCC), differentiation of oncocytoma from RCC, differentiation of different subtypes of RCC, to predict Fuhrman grade, to predict gene mutation through molecular biomarkers and to predict treatment response in metastatic RCC undergoing immunotherapy. Neural networks analyze imaging data. Statistical, geometrical, textural features derived are giving quantitative data of contour, internal heterogeneity and gray zone features of lesions. A comprehensive literature review was performed, until July 2022. Studies investigating the diagnostic value of radiomics in differentiation of renal lesions, grade prediction, gene alterations, molecular biomarkers and ongoing clinical trials have been analyzed. The application of AI and radiomics could lead to improved sensitivity, specificity, accuracy in detecting and differentiating between renal lesions. Standardization of scanner protocols will improve preoperative differentiation between benign, low-risk cancers and clinically significant renal cancers and holds the premises to enhance the diagnostic ability of imaging tools to characterize renal lesions.
Collapse
Affiliation(s)
| | - Felice Crocetto
- Department of Neurosciences and Reproductive
Sciences and Odontostomatology, University of Naples Federico II, Naples,
Italy
| | - Biagio Barone
- Department of Neurosciences and Reproductive
Sciences and Odontostomatology, University of Naples Federico II, Naples,
Italy
| | - Francesco del Giudice
- Department of Maternal Infant and Urologic
Sciences, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome,
Italy
| | - Martina Maggi
- Department of Maternal Infant and Urologic
Sciences, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome,
Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Kidney Transplantation
Unit, Department of Emergency and Organ Transplantation, University of Bari,
Bari, Italy
| | - Gian Maria Busetto
- Department of Urology and Organ
Transplantation, University of Foggia, Foggia, Italy
| | | | - Michele Marchioni
- Department of Medical, Oral and
Biotechnological Sciences, Urology Unit, SS Annunziata Hospital, G.
d’Annunzio University of Chieti, Chieti, Italy
- Department of Urology, ASL Abruzzo 2, Chieti,
Italy
| | - Francesco Cantiello
- Department of Urology, Magna Graecia
University of Catanzaro, Catanzaro, Italy
| | - Fabio Crocerossa
- Department of Urology, Magna Graecia
University of Catanzaro, Catanzaro, Italy
| | - Stefano Luzzago
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
- Università degli Studi di Milano, Milan,
Italy
| | - Mattia Piccinelli
- Cancer Prognostics and Health Outcomes Unit,
Division of Urology, University of Montréal Health Center, Montréal, QC,
Canada
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
| | - Francesco Alessandro Mistretta
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
- Department of Oncology and
Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Marco Tozzi
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
- Università degli Studi di Milano, Milan,
Italy
| | - Luigi Schips
- Department of Medical, Oral and
Biotechnological Sciences, Urology Unit, SS Annunziata Hospital, G.
d’Annunzio University of Chieti, Chieti, Italy
| | | | - Alessandro Veccia
- Urology Unit, Azienda Ospedaliera
Universitaria Integrata Verona, University of Verona, Verona, Italy
| | - Mihai Dorin Vartolomei
- Department of Cell and Molecular Biology,
George Emil Palade University of Medicine, Pharmacy, Science and Technology
of Târgu Mures, Târgu Mures, Romania
- Department of Urology, Medical University of
Vienna, Vienna, Austria
| | - Gennaro Musi
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
- Department of Oncology and
Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Ottavio de Cobelli
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
- Department of Oncology and
Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Emanuele Montanari
- Department of Urology, Foundation IRCCS Ca’
Granda – Ospedale Maggiore Policlinico, Department of Clinical Sciences and
Community Health, University of Milan, Milan, Italy
| | - Octavian Sabin Tătaru
- Institution Organizing University Doctoral
Studies (IOSUD), George Emil Palade University of Medicine, Pharmacy,
Science and Technology of Târgu Mures, Târgu Mures, Romania
| |
Collapse
|
43
|
Cè M, Caloro E, Pellegrino ME, Basile M, Sorce A, Fazzini D, Oliva G, Cellina M. Artificial intelligence in breast cancer imaging: risk stratification, lesion detection and classification, treatment planning and prognosis-a narrative review. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:795-816. [PMID: 36654817 PMCID: PMC9834285 DOI: 10.37349/etat.2022.00113] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/28/2022] [Indexed: 12/28/2022] Open
Abstract
The advent of artificial intelligence (AI) represents a real game changer in today's landscape of breast cancer imaging. Several innovative AI-based tools have been developed and validated in recent years that promise to accelerate the goal of real patient-tailored management. Numerous studies confirm that proper integration of AI into existing clinical workflows could bring significant benefits to women, radiologists, and healthcare systems. The AI-based approach has proved particularly useful for developing new risk prediction models that integrate multi-data streams for planning individualized screening protocols. Furthermore, AI models could help radiologists in the pre-screening and lesion detection phase, increasing diagnostic accuracy, while reducing workload and complications related to overdiagnosis. Radiomics and radiogenomics approaches could extrapolate the so-called imaging signature of the tumor to plan a targeted treatment. The main challenges to the development of AI tools are the huge amounts of high-quality data required to train and validate these models and the need for a multidisciplinary team with solid machine-learning skills. The purpose of this article is to present a summary of the most important AI applications in breast cancer imaging, analyzing possible challenges and new perspectives related to the widespread adoption of these new tools.
Collapse
Affiliation(s)
- Maurizio Cè
- Postgraduate School in Diagnostic and Interventional Radiology, University of Milan, 20122 Milan, Italy
| | - Elena Caloro
- Postgraduate School in Diagnostic and Interventional Radiology, University of Milan, 20122 Milan, Italy
| | - Maria E. Pellegrino
- Postgraduate School in Diagnostic and Interventional Radiology, University of Milan, 20122 Milan, Italy
| | - Mariachiara Basile
- Postgraduate School in Diagnostic and Interventional Radiology, University of Milan, 20122 Milan, Italy
| | - Adriana Sorce
- Postgraduate School in Diagnostic and Interventional Radiology, University of Milan, 20122 Milan, Italy
| | | | - Giancarlo Oliva
- Department of Radiology, ASST Fatebenefratelli Sacco, 20121 Milan, Italy
| | - Michaela Cellina
- Department of Radiology, ASST Fatebenefratelli Sacco, 20121 Milan, Italy
| |
Collapse
|
44
|
Development and validation of a [18F]FDG PET/CT-based radiomics nomogram to predict the prognostic risk of pretreatment diffuse large B cell lymphoma patients. Eur Radiol 2022; 33:3354-3365. [PMID: 36547676 PMCID: PMC10121518 DOI: 10.1007/s00330-022-09301-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/03/2022] [Accepted: 11/13/2022] [Indexed: 12/24/2022]
Abstract
Abstract
Objective
In this study, based on PET/CT radiomics features, we developed and validated a nomogram to predict progression-free survival (PFS) for cases with diffuse large B cell lymphoma (DLBCL) treated with immunochemotherapy.
Methods
This study retrospectively recruited 129 cases with DLBCL. Among them, PET/CT scans were conducted and baseline images were collected for radiomics features along with their clinicopathological features. Radiomics features related to recurrence were screened for survival analysis using univariate Cox regression analysis with p < 0.05. Next, a weighted Radiomics-score (Rad-score) was generated and independent risk factors were obtained from univariate and multivariate Cox regressions to build the nomogram. Furthermore, the nomogram was tested for their ability to predict PFS using time-dependent receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA).
Results
Blood platelet, Rad-score, and gender were included in the nomogram as independent DLBCL risk factors for PFS. We found that the training cohort areas under the curve (AUCs) were 0.79, 0.84, and 0.88, and validation cohort AUCs were 0.67, 0.83, and 0.72, respectively. Further, the DCA and calibration curves confirmed the predictive nomogram’s clinical relevance.
Conclusion
Using Rad-score, blood platelet, and gender of the DLBCL patients, a PET/CT radiomics-based nomogram was developed to guide cases’ recurrence risk assessment prior to treatment. The developed nomogram can help provide more appropriate treatment plans to the cases.
Key Points
• DLBCL cases can be classified into low- and high-risk groups using PET/CT radiomics based Rad-score.
• When combined with other clinical characteristics (gender and blood platelet count), Rad-score can be used to predict the outcome of the pretreatment of DLBCL cases with a certain degree of accuracy.
• A prognostic nomogram was established in this study in order to aid in assessing prognostic risk and providing more accurate treatment plans for DLBCL cases.
Collapse
|
45
|
Fahmy D, Alksas A, Elnakib A, Mahmoud A, Kandil H, Khalil A, Ghazal M, van Bogaert E, Contractor S, El-Baz A. The Role of Radiomics and AI Technologies in the Segmentation, Detection, and Management of Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14246123. [PMID: 36551606 PMCID: PMC9777232 DOI: 10.3390/cancers14246123] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary hepatic neoplasm. Thanks to recent advances in computed tomography (CT) and magnetic resonance imaging (MRI), there is potential to improve detection, segmentation, discrimination from HCC mimics, and monitoring of therapeutic response. Radiomics, artificial intelligence (AI), and derived tools have already been applied in other areas of diagnostic imaging with promising results. In this review, we briefly discuss the current clinical applications of radiomics and AI in the detection, segmentation, and management of HCC. Moreover, we investigate their potential to reach a more accurate diagnosis of HCC and to guide proper treatment planning.
Collapse
Affiliation(s)
- Dalia Fahmy
- Diagnostic Radiology Department, Mansoura University Hospital, Mansoura 35516, Egypt
| | - Ahmed Alksas
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
| | - Ahmed Elnakib
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
| | - Ali Mahmoud
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
| | - Heba Kandil
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
- Faculty of Computer Sciences and Information, Mansoura University, Mansoura 35516, Egypt
| | - Ashraf Khalil
- College of Technological Innovation, Zayed University, Abu Dhabi 4783, United Arab Emirates
| | - Mohammed Ghazal
- Electrical, Computer, and Biomedical Engineering Department, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates
| | - Eric van Bogaert
- Department of Radiology, University of Louisville, Louisville, KY 40202, USA
| | - Sohail Contractor
- Department of Radiology, University of Louisville, Louisville, KY 40202, USA
| | - Ayman El-Baz
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
- Correspondence:
| |
Collapse
|
46
|
Khanna NN, Maindarkar MA, Viswanathan V, Fernandes JFE, Paul S, Bhagawati M, Ahluwalia P, Ruzsa Z, Sharma A, Kolluri R, Singh IM, Laird JR, Fatemi M, Alizad A, Saba L, Agarwal V, Sharma A, Teji JS, Al-Maini M, Rathore V, Naidu S, Liblik K, Johri AM, Turk M, Mohanty L, Sobel DW, Miner M, Viskovic K, Tsoulfas G, Protogerou AD, Kitas GD, Fouda MM, Chaturvedi S, Kalra MK, Suri JS. Economics of Artificial Intelligence in Healthcare: Diagnosis vs. Treatment. Healthcare (Basel) 2022; 10:2493. [PMID: 36554017 PMCID: PMC9777836 DOI: 10.3390/healthcare10122493] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Motivation: The price of medical treatment continues to rise due to (i) an increasing population; (ii) an aging human growth; (iii) disease prevalence; (iv) a rise in the frequency of patients that utilize health care services; and (v) increase in the price. Objective: Artificial Intelligence (AI) is already well-known for its superiority in various healthcare applications, including the segmentation of lesions in images, speech recognition, smartphone personal assistants, navigation, ride-sharing apps, and many more. Our study is based on two hypotheses: (i) AI offers more economic solutions compared to conventional methods; (ii) AI treatment offers stronger economics compared to AI diagnosis. This novel study aims to evaluate AI technology in the context of healthcare costs, namely in the areas of diagnosis and treatment, and then compare it to the traditional or non-AI-based approaches. Methodology: PRISMA was used to select the best 200 studies for AI in healthcare with a primary focus on cost reduction, especially towards diagnosis and treatment. We defined the diagnosis and treatment architectures, investigated their characteristics, and categorized the roles that AI plays in the diagnostic and therapeutic paradigms. We experimented with various combinations of different assumptions by integrating AI and then comparing it against conventional costs. Lastly, we dwell on three powerful future concepts of AI, namely, pruning, bias, explainability, and regulatory approvals of AI systems. Conclusions: The model shows tremendous cost savings using AI tools in diagnosis and treatment. The economics of AI can be improved by incorporating pruning, reduction in AI bias, explainability, and regulatory approvals.
Collapse
Affiliation(s)
- Narendra N. Khanna
- Department of Cardiology, Indraprastha APOLLO Hospitals, New Delhi 110001, India
| | - Mahesh A. Maindarkar
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India
| | | | | | - Sudip Paul
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India
| | - Mrinalini Bhagawati
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India
| | - Puneet Ahluwalia
- Max Institute of Cancer Care, Max Super Specialty Hospital, New Delhi 110017, India
| | - Zoltan Ruzsa
- Invasive Cardiology Division, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Aditya Sharma
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA 22904, USA
| | - Raghu Kolluri
- Ohio Health Heart and Vascular, Columbus, OH 43214, USA
| | - Inder M. Singh
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
| | - John R. Laird
- Heart and Vascular Institute, Adventist Health St. Helena, St. Helena, CA 94574, USA
| | - Mostafa Fatemi
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Azra Alizad
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, 40138 Cagliari, Italy
| | - Vikas Agarwal
- Department of Immunology, SGPGIMS, Lucknow 226014, India
| | - Aman Sharma
- Department of Immunology, SGPGIMS, Lucknow 226014, India
| | - Jagjit S. Teji
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Mustafa Al-Maini
- Allergy, Clinical Immunology and Rheumatology Institute, Toronto, ON L4Z 4C4, Canada
| | | | - Subbaram Naidu
- Electrical Engineering Department, University of Minnesota, Duluth, MN 55812, USA
| | - Kiera Liblik
- Department of Medicine, Division of Cardiology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Amer M. Johri
- Department of Medicine, Division of Cardiology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Monika Turk
- The Hanse-Wissenschaftskolleg Institute for Advanced Study, 27753 Delmenhorst, Germany
| | - Lopamudra Mohanty
- Department of Computer Science, ABES Engineering College, Ghaziabad 201009, India
| | - David W. Sobel
- Rheumatology Unit, National Kapodistrian University of Athens, 15772 Athens, Greece
| | - Martin Miner
- Men’s Health Centre, Miriam Hospital Providence, Providence, RI 02906, USA
| | - Klaudija Viskovic
- Department of Radiology and Ultrasound, University Hospital for Infectious Diseases, 10000 Zagreb, Croatia
| | - George Tsoulfas
- Department of Surgery, Aristoteleion University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Athanasios D. Protogerou
- Cardiovascular Prevention and Research Unit, Department of Pathophysiology, National & Kapodistrian University of Athens, 15772 Athens, Greece
| | - George D. Kitas
- Academic Affairs, Dudley Group NHS Foundation Trust, Dudley DY1 2HQ, UK
- Arthritis Research UK Epidemiology Unit, Manchester University, Manchester M13 9PL, UK
| | - Mostafa M. Fouda
- Department of Electrical and Computer Engineering, Idaho State University, Pocatello, ID 83209, USA
| | - Seemant Chaturvedi
- Department of Neurology & Stroke Program, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Jasjit S. Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
| |
Collapse
|
47
|
Cao R, Tang L, Fang M, Zhong L, Wang S, Gong L, Li J, Dong D, Tian J. Artificial intelligence in gastric cancer: applications and challenges. Gastroenterol Rep (Oxf) 2022; 10:goac064. [PMID: 36457374 PMCID: PMC9707405 DOI: 10.1093/gastro/goac064] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/27/2022] [Accepted: 10/18/2022] [Indexed: 08/10/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors with high mortality. Accurate diagnosis and treatment decisions for GC rely heavily on human experts' careful judgments on medical images. However, the improvement of the accuracy is hindered by imaging conditions, limited experience, objective criteria, and inter-observer discrepancies. Recently, the developments of machine learning, especially deep-learning algorithms, have been facilitating computers to extract more information from data automatically. Researchers are exploring the far-reaching applications of artificial intelligence (AI) in various clinical practices, including GC. Herein, we aim to provide a broad framework to summarize current research on AI in GC. In the screening of GC, AI can identify precancerous diseases and assist in early cancer detection with endoscopic examination and pathological confirmation. In the diagnosis of GC, AI can support tumor-node-metastasis (TNM) staging and subtype classification. For treatment decisions, AI can help with surgical margin determination and prognosis prediction. Meanwhile, current approaches are challenged by data scarcity and poor interpretability. To tackle these problems, more regulated data, unified processing procedures, and advanced algorithms are urgently needed to build more accurate and robust AI models for GC.
Collapse
Affiliation(s)
| | | | - Mengjie Fang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China
| | - Lianzhen Zhong
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
| | - Siwen Wang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, P. R. China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P. R. China
| | - Lixin Gong
- College of Medicine and Biological Information Engineering School, Northeastern University, Shenyang, Liaoning, P. R. China
| | - Jiazheng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, Beijing, P. R. China
| | - Di Dong
- Corresponding authors. Di Dong, CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Beijing 100190, P. R. China. Tel: +86-13811833760; ; Jie Tian, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China. Tel: +86-10-82618465;
| | - Jie Tian
- Corresponding authors. Di Dong, CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Beijing 100190, P. R. China. Tel: +86-13811833760; ; Jie Tian, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, P. R. China. Tel: +86-10-82618465;
| |
Collapse
|
48
|
Chen Y, Tang W, Liu W, Li R, Wang Q, Shen X, Gong J, Gu Y, Peng W. Multiparametric
MR
Imaging Radiomics Signatures for Assessing the Recurrence Risk of
ER
+/
HER2
− Breast Cancer Quantified With 21‐Gene Recurrence Score. J Magn Reson Imaging 2022. [DOI: 10.1002/jmri.28547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Yang Chen
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Wei Tang
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Wei Liu
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Ruimin Li
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Qifeng Wang
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
- Department of Pathology Fudan University Shanghai Cancer Center Shanghai China
| | - Xigang Shen
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Jing Gong
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Yajia Gu
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| | - Weijun Peng
- Department of Radiology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology, Shanghai Medical College Fudan University Shanghai China
| |
Collapse
|
49
|
Artificial Intelligence in Lung Cancer Imaging: Unfolding the Future. Diagnostics (Basel) 2022; 12:diagnostics12112644. [PMID: 36359485 PMCID: PMC9689810 DOI: 10.3390/diagnostics12112644] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/30/2022] Open
Abstract
Lung cancer is one of the malignancies with higher morbidity and mortality. Imaging plays an essential role in each phase of lung cancer management, from detection to assessment of response to treatment. The development of imaging-based artificial intelligence (AI) models has the potential to play a key role in early detection and customized treatment planning. Computer-aided detection of lung nodules in screening programs has revolutionized the early detection of the disease. Moreover, the possibility to use AI approaches to identify patients at risk of developing lung cancer during their life can help a more targeted screening program. The combination of imaging features and clinical and laboratory data through AI models is giving promising results in the prediction of patients’ outcomes, response to specific therapies, and risk for toxic reaction development. In this review, we provide an overview of the main imaging AI-based tools in lung cancer imaging, including automated lesion detection, characterization, segmentation, prediction of outcome, and treatment response to provide radiologists and clinicians with the foundation for these applications in a clinical scenario.
Collapse
|
50
|
Wang S, Lin C, Kolomaya A, Ostdiek-Wille GP, Wong J, Cheng X, Lei Y, Liu C. Compute Tomography Radiomics Analysis on Whole Pancreas Between Healthy Individual and Pancreatic Ductal Adenocarcinoma Patients: Uncertainty Analysis and Predictive Modeling. Technol Cancer Res Treat 2022; 21:15330338221126869. [PMID: 36184987 PMCID: PMC9530578 DOI: 10.1177/15330338221126869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Radiomics is a rapidly growing field that quantitatively extracts image features
in a high-throughput manner from medical imaging. In this study, we analyzed the
radiomics features of the whole pancreas between healthy individuals and
pancreatic cancer patients, and we established a predictive model that can
distinguish cancer patients from healthy individuals based on these radiomics
features. Methods: We retrospectively collected venous-phase scans
of contrast-enhanced computed tomography (CT) images from 181 control subjects
and 85 cancer case subjects for radiomics analysis and predictive modeling. An
attending radiation oncologist delineated the pancreas for all the subjects in
the Varian Eclipse system, and we extracted 924 radiomics features using
PyRadiomics. We established a feature selection pipeline to exclude redundant or
unstable features. We randomly selected 189 cases (60 cancer and 129 control) as
the training set. The remaining 77 subjects (25 cancer and 52 control) as a test
set. We trained a Random Forest model utilizing the stable features to
distinguish the cancer patients from the healthy individuals on the training
dataset. We analyzed the performance of our best model by running 5-fold
cross-validations on the training dataset and applied our best model to the test
set. Results: We identified that 91 radiomics features are stable
against various uncertainty sources, including bin width, resampling, image
transformation, image noise, and segmentation uncertainty. Eight of the 91
features are nonredundant. Our final predictive model, using these 8 features,
has achieved a mean area under the receiver operating characteristic curve (AUC)
of 0.99 ± 0.01 on the training dataset (189 subjects) by cross-validation. The
model achieved an AUC of 0.910 on the independent test set (77 subjects) and an
accuracy of 0.935. Conclusion: CT-based radiomics analysis based on
the whole pancreas can distinguish cancer patients from healthy individuals, and
it could potentially become an early detection tool for pancreatic cancer.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Radiation Oncology, University of Nebraska Medical
Center, Omaha, NE, USA,Shuo Wang, PhD, Department of Radiation
Oncology, University of Nebraska Medical Center, 986861 Nebraska Medical Center,
Omaha, NE 68198, USA. Chi Lin, MD, PhD,
Department of Radiation Oncology, University of Nebraska Medical Center, 986861
Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical
Center, Omaha, NE, USA
| | - Alexander Kolomaya
- College of Medicine, University of Nebraska Medical
Center, Omaha, NE, USA
| | | | - Jeffrey Wong
- Department of Radiation Oncology, University of Nebraska Medical
Center, Omaha, NE, USA
| | - Xiaoyue Cheng
- Department of Mathematics, University of Nebraska
Omaha, Omaha, NE, USA
| | - Yu Lei
- Department of Radiation Oncology, Barrow Neurological
Institute, Phoenix, AZ, USA
| | | |
Collapse
|