1
|
Liao H, Huang C, Liu C, Zhang J, Tao F, Liu H, Liang H, Hu X, Li Y, Chen S, Li Y. Deep learning-based MVIT-MLKA model for accurate classification of pancreatic lesions: a multicenter retrospective cohort study. LA RADIOLOGIA MEDICA 2025:10.1007/s11547-025-01949-5. [PMID: 39832039 DOI: 10.1007/s11547-025-01949-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Accurate differentiation between benign and malignant pancreatic lesions is critical for effective patient management. This study aimed to develop and validate a novel deep learning network using baseline computed tomography (CT) images to predict the classification of pancreatic lesions. METHODS This retrospective study included 864 patients (422 men, 442 women) with confirmed histopathological results across three medical centers, forming a training cohort, internal testing cohort, and external validation cohort. A novel hybrid model, Multi-Scale Large Kernel Attention with Mobile Vision Transformer (MVIT-MLKA), was developed, integrating CNN and Transformer architectures to classify pancreatic lesions. The model's performance was compared with traditional machine learning methods and advanced deep learning models. We also evaluated the diagnostic accuracy of radiologists with and without the assistance of the optimal model. Model performance was assessed through discrimination, calibration, and clinical applicability. RESULTS The MVIT-MLKA model demonstrated superior performance in classifying pancreatic lesions, achieving an AUC of 0.974 (95% CI 0.967-0.980) in the training set, 0.935 (95% CI 0.915-0.954) in the internal testing set, and 0.924 (95% CI 0.902-0.945) in the external validation set, outperforming traditional models and other deep learning models (P < 0.05). Radiologists aided by the MVIT-MLKA model showed significant improvements in diagnostic accuracy and sensitivity compared to those without model assistance (P < 0.05). Grad-CAM visualization enhanced model interpretability by effectively highlighting key lesion areas. CONCLUSION The MVIT-MLKA model efficiently differentiates between benign and malignant pancreatic lesions, surpassing traditional methods and significantly improving radiologists' diagnostic performance. The integration of this advanced deep learning model into clinical practice has the potential to reduce diagnostic errors and optimize treatment strategies.
Collapse
Affiliation(s)
- Hongfan Liao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Cheng Huang
- College of Computer and Information Science, Southwest University, Chongqing, 400715, China
| | - Chunhua Liu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jiao Zhang
- Department of Radiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fengming Tao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Haotian Liu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hongwei Liang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoli Hu
- Department of Radiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Li
- Department of Radiology, The Third People's Hospital of Chengdu, Chengdu, China
| | - Shanxiong Chen
- College of Computer and Information Science, Southwest University, Chongqing, 400715, China.
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
2
|
Zhao B, Cao B, Xia T, Zhu L, Yu Y, Lu C, Tang T, Wang Y, Ju S. Multiparametric MRI for Assessment of the Biological Invasiveness and Prognosis of Pancreatic Ductal Adenocarcinoma in the Era of Artificial Intelligence. J Magn Reson Imaging 2025. [PMID: 39781607 DOI: 10.1002/jmri.29708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/12/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the deadliest malignant tumor, with a grim 5-year overall survival rate of about 12%. As its incidence and mortality rates rise, it is likely to become the second-leading cause of cancer-related death. The radiological assessment determined the stage and management of PDAC. However, it is a highly heterogeneous disease with the complexity of the tumor microenvironment, and it is challenging to adequately reflect the biological aggressiveness and prognosis accurately through morphological evaluation alone. With the dramatic development of artificial intelligence (AI), multiparametric magnetic resonance imaging (mpMRI) using specific contrast media and special techniques can provide morphological and functional information with high image quality and become a powerful tool in quantifying intratumor characteristics. Besides, AI has been widespread in the field of medical imaging analysis. Radiomics is the high-throughput mining of quantitative image features from medical imaging that enables data to be extracted and applied for better decision support. Deep learning is a subset of artificial neural network algorithms that can automatically learn feature representations from data. AI-enabled imaging biomarkers of mpMRI have enormous promise to bridge the gap between medical imaging and personalized medicine and demonstrate huge advantages in predicting biological characteristics and the prognosis of PDAC. However, current AI-based models of PDAC operate mainly in the realm of a single modality with a relatively small sample size, and the technical reproducibility and biological interpretation present a barrage of new potential challenges. In the future, the integration of multi-omics data, such as radiomics and genomics, alongside the establishment of standardized analytical frameworks will provide opportunities to increase the robustness and interpretability of AI-enabled image biomarkers and bring these biomarkers closer to clinical practice. EVIDENCE LEVEL: 3 TECHNICAL EFFICACY: Stage 4.
Collapse
Affiliation(s)
- Ben Zhao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Buyue Cao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Tianyi Xia
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Liwen Zhu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Yaoyao Yu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Chunqiang Lu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Tianyu Tang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Yuancheng Wang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| |
Collapse
|
3
|
Lyu H, Kong J, Chen J, Zhang R, Xiao S, Guo D, Zhang Q, Chen XZ, Tang J, Zhou C. The Emerging Scenario of Ferroptosis in Pancreatic Cancer Tumorigenesis and Treatment. Int J Mol Sci 2024; 25:13334. [PMID: 39769097 PMCID: PMC11727763 DOI: 10.3390/ijms252413334] [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: 11/25/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 01/05/2025] Open
Abstract
Pancreatic cancer remains one of the most lethal forms of cancer. Currently, there is a lack of effective drug treatments for pancreatic cancer. However, as a newly discovered form of non-apoptotic cell death, ferroptosis has garnered increasing attention in relation to pancreatic cancer. Understanding the role of ferroptosis in the tumorigenesis and treatment of pancreatic cancer may enable more effective clinical trials and treatments for pancreatic cancer and may minimize side effects or restrict the emergence of drug resistance. In this review, we summarize the current knowledge regarding the process and underlying mechanisms of ferroptosis, as well as its dual role in both promoting tumorigenesis and facilitating treatment strategies for pancreatic cancer. Additionally, how ferroptosis is implicated in the development of pancreatitis and insulin resistance, indicating that ferroptosis may play an important role in the risk of pancreatitis- and insulin-resistance-related pancreatic cancers, is also addressed.
Collapse
Affiliation(s)
- Hao Lyu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Jinghua Kong
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Jiasi Chen
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Rui Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Shuai Xiao
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Dong Guo
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Qi Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jingfeng Tang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Cefan Zhou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| |
Collapse
|
4
|
Aouad T, Laurent V, Levant P, Rode A, Brillat-Savarin N, Gaillot P, Hoeffel C, Frampas E, Barat M, Russo R, Wagner M, Zappa M, Ernst O, Delagnes A, Fillias Q, Dawi L, Savoye-Collet C, Copin P, Calame P, Reizine E, Luciani A, Bellin MF, Talbot H, Lassau N. Detection and characterization of pancreatic lesion with artificial intelligence: The SFR 2023 artificial intelligence data challenge. Diagn Interv Imaging 2024; 105:395-399. [PMID: 39048455 DOI: 10.1016/j.diii.2024.07.002] [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: 06/19/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
PURPOSE The purpose of the 2023 SFR data challenge was to invite researchers to develop artificial intelligence (AI) models to identify the presence of a pancreatic mass and distinguish between benign and malignant pancreatic masses on abdominal computed tomography (CT) examinations. MATERIALS AND METHODS Anonymized abdominal CT examinations acquired during the portal venous phase were collected from 18 French centers. Abdominal CT examinations were divided into three groups including CT examinations with no lesion, CT examinations with benign pancreatic mass, or CT examinations with malignant pancreatic mass. Each team included at least one radiologist, one data scientist, and one engineer. Pancreatic lesions were annotated by expert radiologists. CT examinations were distributed in balanced batches via a Health Data Hosting certified platform. Data were distributed into four batches, two for training, one for internal evaluation, and one for the external evaluation. Training used 83 % of the data from 14 centers and external evaluation used data from the other four centers. The metric (i.e., final score) used to rank the participants was a weighted average of mean sensitivity, mean precision and mean area under the curve. RESULTS A total of 1037 abdominal CT examinations were divided into two training sets (including 500 and 232 CT examinations), an internal evaluation set (including 139 CT examinations), and an external evaluation set (including 166 CT examinations). The training sets were distributed on September 7 and October 13, 2023, and evaluation sets on October 15, 2023. Ten teams with a total of 93 members participated to the data challenge, with the best final score being 0.72. CONCLUSION This SFR 2023 data challenge based on multicenter CT data suggests that the use of AI for pancreatic lesions detection is possible on real data, but the distinction between benign and malignant pancreatic lesions remains challenging.
Collapse
Affiliation(s)
- Theodore Aouad
- CentraleSupelec, INRIA, CVN, Université Paris-Saclay, 91190 Gif-sur-Yvette, France.
| | - Valerie Laurent
- Department of Radiology, University Hospital of Nancy, Laboratoire IADI INSERM U 1254, 54035 Nancy, France
| | - Paul Levant
- Société Française de Radiologie, 75013 Paris, France
| | - Agnes Rode
- Department of Diagnostic and Interventional Radiology, Hospices Civils de Lyon, Hôpital de la Croix Rousse, 69317 Lyon, France
| | | | - Pénélope Gaillot
- Department of Diagnostic and Interventional Radiology, Assistance Publique-Hopitaux de Paris, CHU de Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Christine Hoeffel
- Department of Radiology, HMB, CHU Reims, 51100 Reims, France; CReSTIC, Université de Reims-Champagne-Ardenne, UFR Sciences Exactes et Naturelles, 51100 Reims, France
| | - Eric Frampas
- Department of Radiology, Hôtel Dieu, CHU Nantes, 44093 Nantes, France
| | - Maxime Barat
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hopitaux de Paris, 75014 Paris, France; Génomique et Signalisation des Tumeurs Endocrines, Institut Cochin, INSERM U 1016, CNRS UMR8104, 75014 Paris, France; Faculté de Médecine, Université Paris Cité, 75006 Paris, France
| | - Roberta Russo
- Department of Radiology, Hôpital Paul Brousse, Assistance Publique-Hopitaux de Paris, 94800 Villejuif, France
| | - Mathilde Wagner
- Department of Radiology, Assistance Publique-Hopitaux de Paris, Sorbonne Université, Hôpital Universitaire Pitié-Salpêtrière, 75013 Paris, France
| | - Magaly Zappa
- Department of Radiology, Centre Hospitalier de Cayenne, Cayenne 97306, France
| | - Olivier Ernst
- Medical Imaging Department, Lille University Hospital, 59000 Lille, France
| | - Anais Delagnes
- Department of Radiology, CHU Angers, Angers University Hospital, 49933 Angers, France
| | - Quentin Fillias
- Department of Radiology, Hospital Lapeyronie, CHU Montpellier, 34000 Montpellier, France
| | - Lama Dawi
- Department of Radiology, Gustave Roussy, 94805 Villejuif, France
| | - Céline Savoye-Collet
- Department of Radiology, Normandie Université, UNIROUEN, Quantif-LITIS EA 4108, Rouen University Hospital, 76031 Rouen, France
| | - Pauline Copin
- Department of Radiology, Hôpital Beaujon, AP-HP.Nord, 92110 Clichy, France
| | - Paul Calame
- Department of Radiology, University of Bourgogne Franche-Comté, CHU Besançon, 25030 Besançon, France
| | - Edouard Reizine
- Department of Radiology, Hopital Henri Mondor, Assistance Publique-Hopitaux de Paris, University Paris Est Créteil, 94000 Créteil, France
| | - Alain Luciani
- Société Française de Radiologie, 75013 Paris, France; Department of Radiology, Hopital Henri Mondor, Assistance Publique-Hopitaux de Paris, University Paris Est Créteil, 94000 Créteil, France; INSERM, U955, Team 18, 94000 Créteil, France
| | - Marie-France Bellin
- Société Française de Radiologie, 75013 Paris, France; Department of Diagnostic and Interventional Radiology, Assistance Publique-Hopitaux de Paris, CHU de Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Hugues Talbot
- CentraleSupelec, INRIA, CVN, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Nathalie Lassau
- Department of Radiology, Gustave Roussy, 94805 Villejuif, France; Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Inserm, CNRS, CEA, BIOMAPS, UMR 1281, Université Paris-Saclay, Villejuif 94800, France
| |
Collapse
|
5
|
Khatkov IE, Lesko KA, Dubtsova EA, Khomeriki SG, Karnaukhov NS, Vinokurova LV, Shurygina EI, Makarenko NV, Izrailov RE, Savina IV, Salimgereeva DA, Kiriukova MA, Bordin DS. [Possibilities of post-processing of multislice computed tomography results in non-invasive diagnosis of pancreatic fibrosis]. TERAPEVT ARKH 2024; 96:780-789. [PMID: 39404723 DOI: 10.26442/00403660.2024.08.202831] [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: 07/21/2024] [Accepted: 07/21/2024] [Indexed: 01/11/2025]
Abstract
AIM To evaluate the possibilities of post-processing of multidetector computed tomography (CT) results in the non-invasive diagnosis of pancreatic fibrosis (PF). MATERIALS AND METHODS The study included 165 patients aged 57.91±13.5 years who underwent preoperative CT during surgical treatment for chronic pancreatitis and pancreatic cancer from April 2022 to February 2024. The normalized contrast ratios of pancreatic tissue in the pancreatic (NCPP) and venous (NCVP) phases, as well as the contrast ratio (CR) were measured. Pathomorphological assessment of PF performed in tissues outside neoplasm or desmoplastic reaction by the Kloppel and Maillet scale. RESULTS The values of post-processing CT results were compared in groups with different degrees of PF. Mean CR values were significantly higher (p=0.001) in patients with severe PF (CR 1.16±0.65 HU) than in patients with mild PF (CR 0.78±0.31 HU). CR value significant increase (p=0.03) was found in patients with signs of inflammatory changes in the pancreas tissue (CR 1.14±0.6 HU) than in those without them (CR 0.81±0.3 HU). There were no significant differences between the values of NCPP and NCVP, and the degree of PF. CONCLUSION The CR value increased in patients with severe degree of PF. There was a relationship between CR value increase and the radiological density of pancreatic tissue in non-contrast phase and presence of early signs of pancreatic inflammatory changes. Thus, there was a relationship between CT postprocessing results and morphological signs of PF, which can be used for pancreatic fibrosis non-invasive diagnosis and identification of additional signs of early chronic pancreatitis.
Collapse
Affiliation(s)
- I E Khatkov
- Loginov Moscow Clinical Scientific Center
- Russian University of Medicine
| | - K A Lesko
- Loginov Moscow Clinical Scientific Center
| | | | | | | | | | | | | | | | - I V Savina
- Loginov Moscow Clinical Scientific Center
| | | | | | - D S Bordin
- Loginov Moscow Clinical Scientific Center
- Russian University of Medicine
- Tver State Medical University
| |
Collapse
|
6
|
Earl J, Fuentes R, Sanchez MEC, de Paredes AGG, Muñoz M, Sanjuanbenito A, Lobo E, Caminoa A, Rodríguez M, Barreto E, López JV, Ruz-Caracuel I, Durán SL, Olcina JRF, Sánchez BL, Páez SC, Torres A, Blázquez J, Sequeros EV, Carrato A. The Spanish Familial Pancreatic Cancer Registry (PANGENFAM): a decade follow-up of individuals at high-risk for pancreatic cancer. Fam Cancer 2024; 23:383-392. [PMID: 38753287 PMCID: PMC11254983 DOI: 10.1007/s10689-024-00388-x] [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: 12/30/2023] [Accepted: 04/04/2024] [Indexed: 07/18/2024]
Abstract
The Spanish Familial Pancreatic Cancer Registry (PANGENFAM) was established in 2009 and aims to characterize the genotype and phenotype of familial pancreatic cancer (FPC). Furthermore, an early detection screening program for pancreatic ductal adenocarcinoma (PDAC) is provided to healthy high-risk individuals from FPC and hereditary pancreatic cancer families (first-degree relatives). This article describes our experience over the last 10 years in high-risk screening. Hereditary and familial pancreatic cancer families were identified through the oncology and gastroenterology units. High-risk individuals underwent annual screening with endoscopic ultrasound (EUS) and magnetic resonance (MRI) from age 40 or 10 years younger than the youngest affected family member. Results: PANGENFAM has enrolled 290 individuals from 143 families, including 52 PDAC cases and 238 high-risk individuals. All high-risk individuals eligible for screening were offered to enter the surveillance program, with 143 currently participating. Pancreatic abnormalities were detected in 94 individuals (median age 53 years (29-83), with common findings including cystic lesions and inhomogeneous parenchyma. Imaging test concordance was 66%. Surgical intervention was performed in 4 high-risk individuals following highly suspicious lesions detected by imaging. PANGENFAM is a valuable resource for science innovation, such as biobanking, with clinical and imaging data available for analysis. For high-risk families, it may offer a potential for early diagnosis. Collaboration with other national and international registries is needed to increase our understanding of the disease biology and to standardize criteria for inclusion and follow-up, optimizing cost-effectiveness and efficacy.
Collapse
Affiliation(s)
- Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain.
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain.
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain.
| | - Raquel Fuentes
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - María E Castillo Sanchez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
| | - Ana García García de Paredes
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - María Muñoz
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Radiology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alfonso Sanjuanbenito
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Eduardo Lobo
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alejandra Caminoa
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Department of Pathology, Hospital Universitario Ramón y Cajal, 28034, Madrid, Spain
| | - Mercedes Rodríguez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - Emma Barreto
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- University of Alcalá, Madrid, Spain
| | - Jorge Villalón López
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Ignacio Ruz-Caracuel
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Department of Pathology, Hospital Universitario Ramón y Cajal, 28034, Madrid, Spain
| | - Sergio López Durán
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - José Ramón Foruny Olcina
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Bárbara Luna Sánchez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Sonia Camaño Páez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Ana Torres
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Javier Blázquez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Vázquez Sequeros
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - Alfredo Carrato
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- University of Alcalá, Madrid, Spain
- Pancreatic Cancer Europe, Brussels, Belgium
| |
Collapse
|
7
|
Mokhtari A, Casale R, Salahuddin Z, Paquier Z, Guiot T, Woodruff HC, Lambin P, Van Laethem JL, Hendlisz A, Bali MA. Development of Clinical Radiomics-Based Models to Predict Survival Outcome in Pancreatic Ductal Adenocarcinoma: A Multicenter Retrospective Study. Diagnostics (Basel) 2024; 14:712. [PMID: 38611625 PMCID: PMC11011556 DOI: 10.3390/diagnostics14070712] [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: 02/17/2024] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
PURPOSE This multicenter retrospective study aims to identify reliable clinical and radiomic features to build machine learning models that predict progression-free survival (PFS) and overall survival (OS) in pancreatic ductal adenocarcinoma (PDAC) patients. METHODS Between 2010 and 2020 pre-treatment contrast-enhanced CT scans of 287 pathology-confirmed PDAC patients from two sites of the Hopital Universitaire de Bruxelles (HUB) and from 47 hospitals within the HUB network were retrospectively analysed. Demographic, clinical, and survival data were also collected. Gross tumour volume (GTV) and non-tumoral pancreas (RPV) were semi-manually segmented and radiomics features were extracted. Patients from two HUB sites comprised the training dataset, while those from the remaining 47 hospitals of the HUB network constituted the testing dataset. A three-step method was used for feature selection. Based on the GradientBoostingSurvivalAnalysis classifier, different machine learning models were trained and tested to predict OS and PFS. Model performances were assessed using the C-index and Kaplan-Meier curves. SHAP analysis was applied to allow for post hoc interpretability. RESULTS A total of 107 radiomics features were extracted from each of the GTV and RPV. Fourteen subgroups of features were selected: clinical, GTV, RPV, clinical & GTV, clinical & GTV & RPV, GTV-volume and RPV-volume both for OS and PFS. Subsequently, 14 Gradient Boosting Survival Analysis models were trained and tested. In the testing dataset, the clinical & GTV model demonstrated the highest performance for OS (C-index: 0.72) among all other models, while for PFS, the clinical model exhibited a superior performance (C-index: 0.70). CONCLUSIONS An integrated approach, combining clinical and radiomics features, excels in predicting OS, whereas clinical features demonstrate strong performance in PFS prediction.
Collapse
Affiliation(s)
- Ayoub Mokhtari
- Radiology Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Roberto Casale
- Radiology Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Zohaib Salahuddin
- Department of Precision Medicine, GROW—Research Institute for Oncology and Reproduction, Maastricht University, 6220MD Maastricht, The Netherlands
| | - Zelda Paquier
- Medical Physics Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Thomas Guiot
- Medical Physics Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Henry C. Woodruff
- Department of Precision Medicine, GROW—Research Institute for Oncology and Reproduction, Maastricht University, 6220MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, GROW—School for Oncology and Reproduction, Maastricht University Medical Centre+, 6229HX Maastricht, The Netherlands
| | - Philippe Lambin
- Department of Precision Medicine, GROW—Research Institute for Oncology and Reproduction, Maastricht University, 6220MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, GROW—School for Oncology and Reproduction, Maastricht University Medical Centre+, 6229HX Maastricht, The Netherlands
| | - Jean-Luc Van Laethem
- Department of Gastroenterology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Alain Hendlisz
- Department of Gastroenterology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Maria Antonietta Bali
- Radiology Department, Institut Jules Bordet Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| |
Collapse
|
8
|
Choi MH, Yoon SB, Lee YJ, Jung ES, Pak S, Han D, Nickel D. Rim enhancement of pancreatic ductal adenocarcinoma: investigating the relationship with DCE-MRI-based radiomics and next-generation sequencing. Front Oncol 2024; 14:1304187. [PMID: 38525415 PMCID: PMC10959187 DOI: 10.3389/fonc.2024.1304187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/16/2024] [Indexed: 03/26/2024] Open
Abstract
Purpose To identify the clinical and genetic variables associated with rim enhancement of pancreatic ductal adenocarcinoma (PDAC) and to develop a dynamic contrast-enhanced (DCE) MRI-based radiomics model for predicting the genetic status from next-generation sequencing (NGS). Materials and methods Patients with PDAC, who underwent pretreatment pancreatic DCE-MRI between November 2019 and July 2021, were eligible in this prospective study. Two radiologists evaluated presence of rim enhancement in PDAC, a known radiological prognostic indicator, on DCE MRI. NGS was conducted for the tissue from the lesion. The Mann-Whitney U and Chi-square tests were employed to identify clinical and genetic variables associated with rim enhancement in PDAC. For continuous variables predicting rim enhancement, the cutoff value was set based on the Youden's index from the receiver operating characteristic (ROC) curve. Radiomics features were extracted from a volume-of-interest of PDAC on four DCE maps (Ktrans, Kep, Ve, and iAUC). A random forest (RF) model was constructed using 10 selected radiomics features from a pool of 392 original features. This model aimed to predict the status of significant NGS variables associated with rim enhancement. The performance of the model was validated using test set. Results A total of 55 patients (32 men; median age 71 years) were randomly assigned to the training (n = 41) and test (n = 14) sets. In the training set, KRAS, TP53, CDKN2A, and SMAD4 mutation rates were 92.3%, 61.8%, 14.5%, and 9.1%, respectively. Tumor size and KRAS variant allele frequency (VAF) differed between rim-enhancing (n = 12) and nonrim-enhancing (n = 29) PDACs with a cutoff of 17.22%. The RF model's average AUC from 10-fold cross-validation for predicting KRAS VAF status was 0.698. In the test set comprising 6 tumors with low KRAS VAF and 8 with high KRAS VAF, the RF model's AUC reached 1.000, achieving a sensitivity of 75.0%, specificity of 100% and accuracy of 87.5%. Conclusion Rim enhancement of PDAC is associated with KRAS VAF derived from NGS-based genetic information. For predicting the KRAS VAF status in PDAC, a radiomics model based on DCE maps showed promising results.
Collapse
Affiliation(s)
- Moon Hyung Choi
- Department of Radiology, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Bae Yoon
- Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Young Joon Lee
- Department of Radiology, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sun Jung
- Department of Hospital Pathology, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seongyong Pak
- Research Collaboration, Siemens Healthineers Ltd., Seoul, Republic of Korea
| | - Dongyeob Han
- Research Collaboration, Siemens Healthineers Ltd., Seoul, Republic of Korea
| | - Dominik Nickel
- MR Applications Predevelopment, Siemens Healthcare GmbH, Erlangen, Germany
| |
Collapse
|
9
|
Anghel C, Grasu MC, Anghel DA, Rusu-Munteanu GI, Dumitru RL, Lupescu IG. Pancreatic Adenocarcinoma: Imaging Modalities and the Role of Artificial Intelligence in Analyzing CT and MRI Images. Diagnostics (Basel) 2024; 14:438. [PMID: 38396476 PMCID: PMC10887967 DOI: 10.3390/diagnostics14040438] [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: 01/10/2024] [Revised: 02/10/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) stands out as the predominant malignant neoplasm affecting the pancreas, characterized by a poor prognosis, in most cases patients being diagnosed in a nonresectable stage. Image-based artificial intelligence (AI) models implemented in tumor detection, segmentation, and classification could improve diagnosis with better treatment options and increased survival. This review included papers published in the last five years and describes the current trends in AI algorithms used in PDAC. We analyzed the applications of AI in the detection of PDAC, segmentation of the lesion, and classification algorithms used in differential diagnosis, prognosis, and histopathological and genomic prediction. The results show a lack of multi-institutional collaboration and stresses the need for bigger datasets in order for AI models to be implemented in a clinically relevant manner.
Collapse
Affiliation(s)
- Cristian Anghel
- Faculty of Medicine, Department of Medical Imaging and Interventional Radiology, Carol Davila University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania; (C.A.); (R.L.D.); (I.G.L.)
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania; (D.A.A.); (G.-I.R.-M.)
| | - Mugur Cristian Grasu
- Faculty of Medicine, Department of Medical Imaging and Interventional Radiology, Carol Davila University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania; (C.A.); (R.L.D.); (I.G.L.)
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania; (D.A.A.); (G.-I.R.-M.)
| | - Denisa Andreea Anghel
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania; (D.A.A.); (G.-I.R.-M.)
| | - Gina-Ionela Rusu-Munteanu
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania; (D.A.A.); (G.-I.R.-M.)
| | - Radu Lucian Dumitru
- Faculty of Medicine, Department of Medical Imaging and Interventional Radiology, Carol Davila University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania; (C.A.); (R.L.D.); (I.G.L.)
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania; (D.A.A.); (G.-I.R.-M.)
| | - Ioana Gabriela Lupescu
- Faculty of Medicine, Department of Medical Imaging and Interventional Radiology, Carol Davila University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania; (C.A.); (R.L.D.); (I.G.L.)
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania; (D.A.A.); (G.-I.R.-M.)
| |
Collapse
|
10
|
Pacella G, Brunese MC, D’Imperio E, Rotondo M, Scacchi A, Carbone M, Guerra G. Pancreatic Ductal Adenocarcinoma: Update of CT-Based Radiomics Applications in the Pre-Surgical Prediction of the Risk of Post-Operative Fistula, Resectability Status and Prognosis. J Clin Med 2023; 12:7380. [PMID: 38068432 PMCID: PMC10707069 DOI: 10.3390/jcm12237380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is the seventh leading cause of cancer-related deaths worldwide. Surgical resection is the main driver to improving survival in resectable tumors, while neoadjuvant treatment based on chemotherapy (and radiotherapy) is the best option-treatment for a non-primally resectable disease. CT-based imaging has a central role in detecting, staging, and managing PDAC. As several authors have proposed radiomics for risk stratification in patients undergoing surgery for PADC, in this narrative review, we have explored the actual fields of interest of radiomics tools in PDAC built on pre-surgical imaging and clinical variables, to obtain more objective and reliable predictors. METHODS The PubMed database was searched for papers published in the English language no earlier than January 2018. RESULTS We found 301 studies, and 11 satisfied our research criteria. Of those included, four were on resectability status prediction, three on preoperative pancreatic fistula (POPF) prediction, and four on survival prediction. Most of the studies were retrospective. CONCLUSIONS It is possible to conclude that many performing models have been developed to get predictive information in pre-surgical evaluation. However, all the studies were retrospective, lacking further external validation in prospective and multicentric cohorts. Furthermore, the radiomics models and the expression of results should be standardized and automatized to be applicable in clinical practice.
Collapse
Affiliation(s)
- Giulia Pacella
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| | - Maria Chiara Brunese
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| | | | - Marco Rotondo
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| | - Andrea Scacchi
- General Surgery Unit, University of Milano-Bicocca, 20126 Milan, Italy
| | - Mattia Carbone
- San Giovanni di Dio e Ruggi d’Aragona Hospital, 84131 Salerno, Italy;
| | - Germano Guerra
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| |
Collapse
|
11
|
Homps M, Soyer P, Coriat R, Dermine S, Pellat A, Fuks D, Marchese U, Terris B, Groussin L, Dohan A, Barat M. A preoperative computed tomography radiomics model to predict disease-free survival in patients with pancreatic neuroendocrine tumors. Eur J Endocrinol 2023; 189:476-484. [PMID: 37787635 DOI: 10.1093/ejendo/lvad130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 10/04/2023]
Abstract
IMPORTANCE Imaging has demonstrated capabilities in the diagnosis of pancreatic neuroendocrine tumors (pNETs), but its utility for prognostic prediction has not been elucidated yet. OBJECTIVE The aim of this study was to build a radiomics model using preoperative computed tomography (CT) data that may help predict recurrence-free survival (RFS) or OS in patients with pNET. DESIGN We performed a retrospective observational study in a cohort of French patients with pNETs. PARTICIPANTS Patients with surgically resected pNET and available CT examinations were included. INTERVENTIONS Radiomics features of preoperative CT data were extracted using 3D-Slicer® software with manual segmentation. Discriminant features were selected with penalized regression using least absolute shrinkage and selection operator method with training on the tumor Ki67 rate (≤2 or >2). Selected features were used to build a radiomics index ranging from 0 to 1. OUTCOME AND MEASURE A receiving operator curve was built to select an optimal cutoff value of the radiomics index to predict patient RFS and OS. Recurrence-free survival and OS were assessed using Kaplan-Meier analysis. RESULTS Thirty-seven patients (median age, 61 years; 20 men) with 37 pNETs (grade 1, 21/37 [57%]; grade 2, 12/37 [32%]; grade 3, 4/37 [11%]) were included. Patients with a radiomics index >0.4 had a shorter median RFS (36 months; range: 1-133) than those with a radiomics index ≤0.4 (84 months; range: 9-148; P = .013). No associations were found between the radiomics index and OS (P = .86).
Collapse
Affiliation(s)
- Margaux Homps
- Department of Diagnostic and Interventional Imaging, APHP, Hôpital Cochin, Paris F-75014, France
- Faculté de Médecine, Université Paris Cité, Paris F-75006, France
| | - Philippe Soyer
- Department of Diagnostic and Interventional Imaging, APHP, Hôpital Cochin, Paris F-75014, France
- Faculté de Médecine, Université Paris Cité, Paris F-75006, France
| | - Romain Coriat
- Faculté de Médecine, Université Paris Cité, Paris F-75006, France
- Department of Gastroenterology and Digestive Oncology, AP-HP, Hôpital Cochin, Paris F-75014, France
| | - Solène Dermine
- Faculté de Médecine, Université Paris Cité, Paris F-75006, France
- Department of Gastroenterology and Digestive Oncology, AP-HP, Hôpital Cochin, Paris F-75014, France
| | - Anna Pellat
- Faculté de Médecine, Université Paris Cité, Paris F-75006, France
- Department of Gastroenterology and Digestive Oncology, AP-HP, Hôpital Cochin, Paris F-75014, France
| | - David Fuks
- Faculté de Médecine, Université Paris Cité, Paris F-75006, France
- Department of Surgery, Hôpital Cochin, APHP, Paris F-75014, France
| | - Ugo Marchese
- Faculté de Médecine, Université Paris Cité, Paris F-75006, France
- Department of Surgery, Hôpital Cochin, APHP, Paris F-75014, France
| | - Benoit Terris
- Faculté de Médecine, Université Paris Cité, Paris F-75006, France
- Department of Pathology, Center for Rare Adrenal Diseases, AP-HP, Hôpital Cochin, Paris F-75014, France
| | - Lionel Groussin
- Faculté de Médecine, Université Paris Cité, Paris F-75006, France
- Department of Endocrinology, Center for Rare Adrenal Diseases, AP-HP, Hôpital Cochin, Paris F-75014, France
| | - Anthony Dohan
- Department of Diagnostic and Interventional Imaging, APHP, Hôpital Cochin, Paris F-75014, France
- Faculté de Médecine, Université Paris Cité, Paris F-75006, France
| | - Maxime Barat
- Department of Diagnostic and Interventional Imaging, APHP, Hôpital Cochin, Paris F-75014, France
- Faculté de Médecine, Université Paris Cité, Paris F-75006, France
| |
Collapse
|
12
|
Malcolm JA, Tacey M, Gibbs P, Lee B, Ko HS. Current state of radiomic research in pancreatic cancer: focusing on study design and reproducibility of findings. Eur Radiol 2023; 33:6659-6669. [PMID: 37079029 PMCID: PMC10511615 DOI: 10.1007/s00330-023-09653-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 04/21/2023]
Abstract
OBJECTIVES To critically appraise methodology and reproducibility of published studies on CT radiomics of pancreatic ductal adenocarcinoma (PDAC). METHODS PRISMA literature search of MEDLINE, PubMed, and Scopus databases was conducted from June to August 2022 relating to CT radiomics human research articles pertaining to PDAC diagnosis, treatment, and/ or prognosis, utilising Image Biomarker Standardisation Initiative-compliant (IBSI) radiomic software. Keyword search included [pancreas OR pancreatic] AND [radiomic OR [quantitative AND imaging] OR [texture AND analysis]]. Analysis included cohort size, CT protocol used, radiomic feature (RF) extraction, segmentation, and selection, software used, outcome correlation, and statistical methodology, with focus on reproducibility. RESULTS Initial search yielded 1112 articles; however, only 12 articles met all inclusion/exclusion criteria. Cohort sizes ranged from 37 to 352 (median = 106, mean = 155.8). CT slice thickness varied among studies (4 using ≤ 1 mm, 5 using > 1 to 3 mm, 2 using > 3 to 5 mm, 1 not specifying). CT protocol varied (5 using a single portal-venous (pv)-phase, 5 using a pancreas protocol, 1 study using a non-contrast protocol). RF extraction and segmentation were heterogeneous (RF extraction: 5 using pv-phase, 2 using late arterial, 4 using multi-phase, 1 using non-contrast phase; RF selection: 3 pre-selected, 9 software-selected). 2D/3D RF segmentation was diverse (2D in 6, 3D in 4, 2D and 3D in 2 studies). Six different radiomics software were used. Research questions and cohort characteristics varied, ultimately leading to non-comparable outcome results. CONCLUSION The current twelve published IBSI-compliant PDAC radiomic studies show high variability and often incomplete methodology resulting in low robustness and reproducibility. CLINICAL RELEVANCE STATEMENT Radiomics research requires IBSI compliance, data harmonisation, and reproducible feature extraction methods for non-invasive imaging biomarker discoveries to be valid. This will ensure a successful clinical implementation and ultimately an improvement of patient outcomes as part of precision and personalised medicine. KEY POINTS • Current state of radiomics research in pancreatic cancer shows low software compliance to the Image Biomarker Standardisation Initiative (IBSI). • IBSI-compliant radiomics studies in pancreatic cancer are heterogeneous and not comparable, and the majority of study designs showed low reproducibility. • Improved methodology and standardisation of practice in the emerging field of radiomics has the potential of this non-invasive imaging biomarker in the management of pancreatic cancer.
Collapse
Affiliation(s)
- James Alex Malcolm
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
- Department of Cancer Imaging, The Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mark Tacey
- Department of Biostatistics, Northern Health, Epping, VIC, Australia
| | - Peter Gibbs
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Oncology, Western Health, Melbourne, VIC, Australia
- Department of Medical Oncology, Melbourne Health, Melbourne, VIC, Australia
| | - Belinda Lee
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Oncology, Melbourne Health, Melbourne, VIC, Australia
- Department of Medical Oncology, The Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Medical Oncology, Northern Health, Epping, VIC, Australia
| | - Hyun Soo Ko
- Department of Cancer Imaging, The Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia.
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
13
|
Pellat A, Barat M. Tumor microenvironment: A new application for radiomics. Diagn Interv Imaging 2023; 104:93-94. [PMID: 36880939 DOI: 10.1016/j.diii.2022.10.011] [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: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022]
Affiliation(s)
- Anna Pellat
- Gastroenterology and Digestive Oncology Unit, Hôpital Cochin, AP-HP Centre, 75014 Paris, France; Université Paris Cité, Faculté de Médecine, 75006 Paris, France.
| | - Maxime Barat
- Université Paris Cité, Faculté de Médecine, 75006 Paris, France; Department of Radiology, Hôpital Cochin, APHP, 75014 Paris, France
| |
Collapse
|
14
|
Granata V, Fusco R, Setola SV, Galdiero R, Maggialetti N, Silvestro L, De Bellis M, Di Girolamo E, Grazzini G, Chiti G, Brunese MC, Belli A, Patrone R, Palaia R, Avallone A, Petrillo A, Izzo F. Risk Assessment and Pancreatic Cancer: Diagnostic Management and Artificial Intelligence. Cancers (Basel) 2023; 15:351. [PMID: 36672301 PMCID: PMC9857317 DOI: 10.3390/cancers15020351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers, and it is responsible for a number of deaths almost equal to its incidence. The high mortality rate is correlated with several explanations; the main one is the late disease stage at which the majority of patients are diagnosed. Since surgical resection has been recognised as the only curative treatment, a PC diagnosis at the initial stage is believed the main tool to improve survival. Therefore, patient stratification according to familial and genetic risk and the creation of screening protocol by using minimally invasive diagnostic tools would be appropriate. Pancreatic cystic neoplasms (PCNs) are subsets of lesions which deserve special management to avoid overtreatment. The current PC screening programs are based on the annual employment of magnetic resonance imaging with cholangiopancreatography sequences (MR/MRCP) and/or endoscopic ultrasonography (EUS). For patients unfit for MRI, computed tomography (CT) could be proposed, although CT results in lower detection rates, compared to MRI, for small lesions. The actual major limit is the incapacity to detect and characterize the pancreatic intraepithelial neoplasia (PanIN) by EUS and MR/MRCP. The possibility of utilizing artificial intelligence models to evaluate higher-risk patients could favour the diagnosis of these entities, although more data are needed to support the real utility of these applications in the field of screening. For these motives, it would be appropriate to realize screening programs in research settings.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 41012 Napoli, Italy
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, 20122 Milan, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Galdiero
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Nicola Maggialetti
- Department of Medical Science, Neuroscience and Sensory Organs (DSMBNOS), University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Lucrezia Silvestro
- Division of Clinical Experimental Oncology Abdomen, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Mario De Bellis
- Division of Gastroenterology and Digestive Endoscopy, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Elena Di Girolamo
- Division of Gastroenterology and Digestive Endoscopy, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Giulia Grazzini
- Department of Emergency Radiology, University Hospital Careggi, Largo Brambilla 3, 50134 Florence, Italy
| | - Giuditta Chiti
- Department of Emergency Radiology, University Hospital Careggi, Largo Brambilla 3, 50134 Florence, Italy
| | - Maria Chiara Brunese
- Diagnostic Imaging Section, Department of Medical and Surgical Sciences & Neurosciences, University of Molise, 86100 Campobasso, Italy
| | - Andrea Belli
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Renato Patrone
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Raffaele Palaia
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Antonio Avallone
- Division of Clinical Experimental Oncology Abdomen, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| |
Collapse
|
15
|
Xu F, Huang M, Bai Y, Yin X, Yan J, Liu F, Chen J, Weng X. Landmarks in pancreatic cancer studies. Cancer Cell Int 2022; 22:383. [PMID: 36476236 PMCID: PMC9730569 DOI: 10.1186/s12935-022-02803-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is a rare but fatal disease. Patients present advanced disease due to the lack of or typical symptoms when the tumor is still localized. A high-quality image processing system has been in practice to detect the pancreatic tumor and determine the possibility of surgery, and preoperative methods, such as ERCP are increasingly used to complement the staging modality. Pancreaticoduodenectomy is one of the complicated surgeries with potential morbidity. The minimally invasive pancreatic resections, both robot-assisted and laparoscopic, have become a part of standard surgical practice worldwide over the last decade. Moreover, advancements in adjuvant chemotherapy have improved the long-term outcomes in current clinical practice. The systemic conservative treatment, including targeted agents, remains the mainstay of treatment for patients with advanced disease. An increasing number of studies are focused on modulating the pancreatic tumor microenvironment to improve the efficacy of the immunotherapeutic strategies. Herein, the role of preoperative therapy, the novel surgical strategy, and individualized systemic treatment in pancreatic cancer is investigated. Also, the randomized controlled studies that have defined the neoadjuvant and surgical management of pancreatic cancer have been summarized.
Collapse
Affiliation(s)
- Fan Xu
- grid.413856.d0000 0004 1799 3643Department of Public Health, Chengdu Medical College, Chengdu, 610500 Sichuan China
| | - Min Huang
- grid.413856.d0000 0004 1799 3643Department of Physiology, Chengdu Medical College, Chengdu, 610500 Sichuan China
| | - Yun Bai
- grid.413856.d0000 0004 1799 3643Department of Public Health, Chengdu Medical College, Chengdu, 610500 Sichuan China
| | - Xueshi Yin
- grid.413856.d0000 0004 1799 3643Department of Clinic Medicine, Chengdu Medical College, Chengdu, 610500 Sichuan China
| | - Jingzhe Yan
- grid.440230.10000 0004 1789 4901Department of Abdominal Oncosurgery-2, Jilin Province Tumor Hospital, Changchun, 130012 China
| | - Fangfang Liu
- grid.412723.10000 0004 0604 889XArt college, Southwest Minzu University, Chengdu, 610041 Sichuan China
| | - Jie Chen
- grid.412277.50000 0004 1760 6738Department of Orthopedics, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China ,grid.194645.b0000000121742757School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, 999077 China
| | - Xiechuan Weng
- grid.506261.60000 0001 0706 7839Department of Neuroscience, Beijing Institute of Basic Medical Sciences, Beijing, 100850 China
| |
Collapse
|
16
|
Liu X, Elbanan MG, Luna A, Haider MA, Smith AD, Sabottke CF, Spieler BM, Turkbey B, Fuentes D, Moawad A, Kamel S, Horvat N, Elsayes KM. Radiomics in Abdominopelvic Solid-Organ Oncologic Imaging: Current Status. AJR Am J Roentgenol 2022; 219:985-995. [PMID: 35766531 PMCID: PMC10616929 DOI: 10.2214/ajr.22.27695] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Radiomics is the process of extraction of high-throughput quantitative imaging features from medical images. These features represent noninvasive quantitative biomarkers that go beyond the traditional imaging features visible to the human eye. This article first reviews the steps of the radiomics pipeline, including image acquisition, ROI selection and image segmentation, image preprocessing, feature extraction, feature selection, and model development and application. Current evidence for the application of radiomics in abdominopelvic solid-organ cancers is then reviewed. Applications including diagnosis, subtype determination, treatment response assessment, and outcome prediction are explored within the context of hepatobiliary and pancreatic cancer, renal cell carcinoma, prostate cancer, gynecologic cancer, and adrenal masses. This literature review focuses on the strongest available evidence, including systematic reviews, meta-analyses, and large multicenter studies. Limitations of the available literature are highlighted, including marked heterogeneity in radiomics methodology, frequent use of small sample sizes with high risk of overfitting, and lack of prospective design, external validation, and standardized radiomics workflow. Thus, although studies have laid a foundation that supports continued investigation into radiomics models, stronger evidence is needed before clinical adoption.
Collapse
Affiliation(s)
- Xiaoyang Liu
- Joint Department of Medical Imaging, Division of Abdominal Imaging, University Health Network, University of Toronto, ON, Canada
| | - Mohamed G Elbanan
- Department of Radiology, Yale New Haven Health, Bridgeport Hospital, Bridgeport, CT
| | | | - Masoom A Haider
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Joint Department of Medical Imaging, University Health Network, Sinai Health System and University of Toronto, Toronto, ON, Canada
| | - Andrew D Smith
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Carl F Sabottke
- Department of Medical Imaging, University of Arizona College of Medicine, Tucson, AZ
| | - Bradley M Spieler
- Department of Radiology, University Medical Center, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, NIH, Bethesda, MD
| | - David Fuentes
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ahmed Moawad
- Department of Diagnostic and Interventional Radiology, Mercy Catholic Medical Center, Darby, PA
| | - Serageldin Kamel
- Department of Lymphoma, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Natally Horvat
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Khaled M Elsayes
- Department of Abdominal Imaging, University of Texas MD Anderson Cancer Center, 1400 Pressler St, Houston, TX 77030
| |
Collapse
|
17
|
Liang W, Tian W, Wang Y, Wang P, Wang Y, Zhang H, Ruan S, Shao J, Zhang X, Huang D, Ding Y, Bai X. Classification prediction of pancreatic cystic neoplasms based on radiomics deep learning models. BMC Cancer 2022; 22:1237. [PMID: 36447168 PMCID: PMC9710154 DOI: 10.1186/s12885-022-10273-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Preoperative prediction of pancreatic cystic neoplasm (PCN) differentiation has significant value for the implementation of personalized diagnosis and treatment plans. This study aimed to build radiomics deep learning (DL) models using computed tomography (CT) data for the preoperative differential diagnosis of common cystic tumors of the pancreas. METHODS Clinical and CT data of 193 patients with PCN were collected for this study. Among these patients, 99 were pathologically diagnosed with pancreatic serous cystadenoma (SCA), 55 were diagnosed with mucinous cystadenoma (MCA) and 39 were diagnosed with intraductal papillary mucinous neoplasm (IPMN). The regions of interest (ROIs) were obtained based on manual image segmentation of CT slices. The radiomics and radiomics-DL models were constructed using support vector machines (SVMs). Moreover, based on the fusion of clinical and radiological features, the best combined feature set was obtained according to the Akaike information criterion (AIC) analysis. Then the fused model was constructed using logistic regression. RESULTS For the SCA differential diagnosis, the fused model performed the best and obtained an average area under the curve (AUC) of 0.916. It had a best feature set including position, polycystic features (≥6), cystic wall calcification, pancreatic duct dilatation and radiomics-DL score. For the MCA and IPMN differential diagnosis, the fused model with AUC of 0.973 had a best feature set including age, communication with the pancreatic duct and radiomics score. CONCLUSIONS The radiomics, radiomics-DL and fused models based on CT images have a favorable differential diagnostic performance for SCA, MCA and IPMN. These findings may be beneficial for the exploration of individualized management strategies.
Collapse
Affiliation(s)
- Wenjie Liang
- grid.13402.340000 0004 1759 700XDepartment of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou China
| | - Wuwei Tian
- grid.13402.340000 0004 1759 700XCollege of Information Science & Electronic Engineering, School of Micro-Nano Electronics, Zhejiang University, Zheda Road, Zhejiang, Hangzhou China
| | - Yifan Wang
- grid.13402.340000 0004 1759 700XCollege of Information Science & Electronic Engineering, School of Micro-Nano Electronics, Zhejiang University, Zheda Road, Zhejiang, Hangzhou China
| | - Pan Wang
- grid.13402.340000 0004 1759 700XDepartment of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou China
| | - Yubizhuo Wang
- grid.13402.340000 0004 1759 700XDepartment of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou China
| | - Hongbin Zhang
- grid.513202.7Department of Radiology, Yiwu Central Hospital, Yiwu, Zhejiang, China
| | - Shijian Ruan
- grid.13402.340000 0004 1759 700XCollege of Information Science & Electronic Engineering, Zhejiang University, Zhejiang, Hangzhou China
| | - Jiayuan Shao
- grid.13402.340000 0004 1759 700XPolytechnic Institute, Zhejiang University, Zhejiang, Hangzhou China
| | - Xiuming Zhang
- grid.13402.340000 0004 1759 700XDepartment of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou China
| | - Danjiang Huang
- grid.469601.cDepartment of Radiology, Taizhou First People’s Hospital, Taizhou, Zhejiang, China
| | - Yong Ding
- grid.13402.340000 0004 1759 700XCollege of Information Science & Electronic Engineering, School of Micro-Nano Electronics, Zhejiang University, Zheda Road, Zhejiang, Hangzhou China
| | - Xueli Bai
- grid.452661.20000 0004 1803 6319Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Qingchun Road, Zhejiang, Hangzhou China ,grid.452661.20000 0004 1803 6319Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou China
| |
Collapse
|
18
|
Phantom Study on the Robustness of MR Radiomics Features: Comparing the Applicability of 3D Printed and Biological Phantoms. Diagnostics (Basel) 2022; 12:diagnostics12092196. [PMID: 36140598 PMCID: PMC9497898 DOI: 10.3390/diagnostics12092196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
The objectives of our study were to (a) evaluate the feasibility of using 3D printed phantoms in magnetic resonance imaging (MR) in assessing the robustness and repeatability of radiomic parameters and (b) to compare the results obtained from the 3D printed phantoms to metrics obtained in biological phantoms. To this end, three different 3D phantoms were printed: a Hilbert cube (5 × 5 × 5 cm3) and two cubic quick response (QR) code phantoms (a large phantom (large QR) (5 × 5 × 4 cm3) and a small phantom (small QR) (4 × 4 × 3 cm3)). All 3D printed and biological phantoms (kiwis, tomatoes, and onions) were scanned thrice on clinical 1.5 T and 3 T MR with 1 mm and 2 mm isotropic resolution. Subsequent analyses included analyses of several radiomics indices (RI), their repeatability and reliability were calculated using the coefficient of variation (CV), the relative percentage difference (RPD), and the interclass coefficient (ICC) parameters. Additionally, the readability of QR codes obtained from the MR images was examined with several mobile phones and algorithms. The best repeatability (CV ≤ 10%) is reported for the acquisition protocols with the highest spatial resolution. In general, the repeatability and reliability of RI were better in data obtained at 1.5 T (CV = 1.9) than at 3 T (CV = 2.11). Furthermore, we report good agreements between results obtained for the 3D phantoms and biological phantoms. Finally, analyses of the read-out rate of the QR code revealed better texture analyses for images with a spatial resolution of 1 mm than 2 mm. In conclusion, 3D printing techniques offer a unique solution to create textures for analyzing the reliability of radiomic data from MR scans.
Collapse
|
19
|
Barat M, Marchese U, Pellat A, Dohan A, Coriat R, Hoeffel C, Fishman EK, Cassinotto C, Chu L, Soyer P. Imaging of Pancreatic Ductal Adenocarcinoma: An Update on Recent Advances. Can Assoc Radiol J 2022; 74:351-361. [PMID: 36065572 DOI: 10.1177/08465371221124927] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Pancreatic ductal carcinoma (PDAC) is one of the leading causes of cancer-related death worldwide. Computed tomography (CT) remains the primary imaging modality for diagnosis of PDAC. However, CT has limitations for early pancreatic tumor detection and tumor characterization so that it is currently challenged by magnetic resonance imaging. More recently, a particular attention has been given to radiomics for the characterization of pancreatic lesions using extraction and analysis of quantitative imaging features. In addition, radiomics has currently many applications that are developed in conjunction with artificial intelligence (AI) with the aim of better characterizing pancreatic lesions and providing a more precise assessment of tumor burden. This review article sums up recent advances in imaging of PDAC in the field of image/data acquisition, tumor detection, tumor characterization, treatment response evaluation, and preoperative planning. In addition, current applications of radiomics and AI in the field of PDAC are discussed.
Collapse
Affiliation(s)
- Maxime Barat
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris543341, Paris, France.,Université Paris Cité, Faculté de Médecine, 555089Paris, France
| | - Ugo Marchese
- Université Paris Cité, Faculté de Médecine, 555089Paris, France.,Department of Digestive, Hepatobiliary and Pancreatic Surgery, 26935Hopital Cochin, AP-HP, Paris, France
| | - Anna Pellat
- Université Paris Cité, Faculté de Médecine, 555089Paris, France.,Department of Gastroenterology, 26935Hopital Cochin, AP-HP, Paris, France
| | - Anthony Dohan
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris543341, Paris, France.,Université Paris Cité, Faculté de Médecine, 555089Paris, France
| | - Romain Coriat
- Université Paris Cité, Faculté de Médecine, 555089Paris, France.,Department of Gastroenterology, 26935Hopital Cochin, AP-HP, Paris, France
| | | | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, 1466Johns Hopkins University, Baltimore, MD, USA
| | - Christophe Cassinotto
- Department of Radiology, CHU Montpellier, 27037University of Montpellier, Saint-Éloi Hospital, Montpellier, France
| | - Linda Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, 1466Johns Hopkins University, Baltimore, MD, USA
| | - Philippe Soyer
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris543341, Paris, France.,Université Paris Cité, Faculté de Médecine, 555089Paris, France
| |
Collapse
|
20
|
Marti-Bonmati L, Cerdá-Alberich L, Pérez-Girbés A, Díaz Beveridge R, Montalvá Orón E, Pérez Rojas J, Alberich-Bayarri A. Pancreatic cancer, radiomics and artificial intelligence. Br J Radiol 2022; 95:20220072. [PMID: 35687700 PMCID: PMC10996946 DOI: 10.1259/bjr.20220072] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/19/2022] [Accepted: 05/27/2022] [Indexed: 11/05/2022] Open
Abstract
Patients with pancreatic ductal adenocarcinoma (PDAC) are generally classified into four categories based on contrast-enhanced CT at diagnosis: resectable, borderline resectable, unresectable, and metastatic disease. In the initial grading and staging of PDAC, structured radiological templates are useful but limited, as there is a need to define the aggressiveness and microscopic disease stage of these tumours to ensure adequate treatment allocation. Quantitative imaging analysis allows radiomics and dynamic imaging features to provide information of clinical outcomes, and to construct clinical models based on radiomics signatures or imaging phenotypes. These quantitative features may be used as prognostic and predictive biomarkers in clinical decision-making, enabling personalised management of advanced PDAC. Deep learning and convolutional neural networks also provide high level bioinformatics tools that can help define features associated with a given aspect of PDAC biology and aggressiveness, paving the way to define outcomes based on these features. Thus, the prediction of tumour phenotype, treatment response and patient prognosis may be feasible by using such comprehensive and integrated radiomics models. Despite these promising results, quantitative imaging is not ready for clinical implementation in PDAC. Limitations include the instability of metrics and lack of external validation. Large properly annotated datasets, including relevant semantic features (demographics, blood markers, genomics), image harmonisation, robust radiomics analysis, clinically significant tasks as outputs, comparisons with gold-standards (such as TNM or pretreatment classifications) and fully independent validation cohorts, will be required for the development of trustworthy radiomics and artificial intelligence solutions to predict PDAC aggressiveness in a clinical setting.
Collapse
Affiliation(s)
- Luis Marti-Bonmati
- GIBI230 Research Group on Biomedical Imaging, Instituto de
Investigación Sanitaria La Fe,
Valencia, Spain
- Department of Radiology, Hospital Universitario y
Politécnico La Fe, Valencia,
Spain
| | - Leonor Cerdá-Alberich
- GIBI230 Research Group on Biomedical Imaging, Instituto de
Investigación Sanitaria La Fe,
Valencia, Spain
| | | | | | - Eva Montalvá Orón
- Department of Surgery, Hospital Universitario y
Politécnico La Fe, Valencia,
Spain
| | - Judith Pérez Rojas
- Department of Pathology, Hospital Universitario y
Politécnico La Fe, Valencia,
Spain
| | - Angel Alberich-Bayarri
- GIBI230 Research Group on Biomedical Imaging, Instituto de
Investigación Sanitaria La Fe,
Valencia, Spain
- Quantitative Imaging Biomarkers in Medicine, Quibim
SL, Valencia,
Spain
| |
Collapse
|
21
|
Lafata KJ, Wang Y, Konkel B, Yin FF, Bashir MR. Radiomics: a primer on high-throughput image phenotyping. Abdom Radiol (NY) 2022; 47:2986-3002. [PMID: 34435228 DOI: 10.1007/s00261-021-03254-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 01/18/2023]
Abstract
Radiomics is a high-throughput approach to image phenotyping. It uses computer algorithms to extract and analyze a large number of quantitative features from radiological images. These radiomic features collectively describe unique patterns that can serve as digital fingerprints of disease. They may also capture imaging characteristics that are difficult or impossible to characterize by the human eye. The rapid development of this field is motivated by systems biology, facilitated by data analytics, and powered by artificial intelligence. Here, as part of Abdominal Radiology's special issue on Quantitative Imaging, we provide an introduction to the field of radiomics. The technique is formally introduced as an advanced application of data analytics, with illustrating examples in abdominal radiology. Artificial intelligence is then presented as the main driving force of radiomics, and common techniques are defined and briefly compared. The complete step-by-step process of radiomic phenotyping is then broken down into five key phases. Potential pitfalls of each phase are highlighted, and recommendations are provided to reduce sources of variation, non-reproducibility, and error associated with radiomics.
Collapse
Affiliation(s)
- Kyle J Lafata
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA. .,Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA. .,Department of Electrical & Computer Engineering, Duke University Pratt School of Engineering, Durham, NC, USA.
| | - Yuqi Wang
- Department of Electrical & Computer Engineering, Duke University Pratt School of Engineering, Durham, NC, USA
| | - Brandon Konkel
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Fang-Fang Yin
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Mustafa R Bashir
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA.,Department of Medicine, Gastroenterology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
22
|
Update on quantitative radiomics of pancreatic tumors. Abdom Radiol (NY) 2022; 47:3118-3160. [PMID: 34292365 DOI: 10.1007/s00261-021-03216-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
Radiomics is a newer approach for analyzing radiological images obtained from conventional imaging modalities such as computed tomography, magnetic resonance imaging, endoscopic ultrasonography, and positron emission tomography. Radiomics involves extracting quantitative data from the images and assessing them to identify diagnostic or prognostic features such as tumor grade, resectability, tumor response to neoadjuvant therapy, and survival. The purpose of this review is to discuss the basic principles of radiomics and provide an overview of the current clinical applications of radiomics in the field of pancreatic tumors.
Collapse
|
23
|
A systematic review of radiomics in pancreatitis: applying the evidence level rating tool for promoting clinical transferability. Insights Imaging 2022; 13:139. [PMID: 35986798 PMCID: PMC9391628 DOI: 10.1186/s13244-022-01279-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/26/2022] [Indexed: 12/16/2022] Open
Abstract
Background Multiple tools have been applied to radiomics evaluation, while evidence rating tools for this field are still lacking. This study aims to assess the quality of pancreatitis radiomics research and test the feasibility of the evidence level rating tool. Results Thirty studies were included after a systematic search of pancreatitis radiomics studies until February 28, 2022, via five databases. Twenty-four studies employed radiomics for diagnostic purposes. The mean ± standard deviation of the adherence rate was 38.3 ± 13.3%, 61.3 ± 11.9%, and 37.1 ± 27.2% for the Radiomics Quality Score (RQS), the Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis (TRIPOD) checklist, and the Image Biomarker Standardization Initiative (IBSI) guideline for preprocessing steps, respectively. The median (range) of RQS was 7.0 (− 3.0 to 18.0). The risk of bias and application concerns were mainly related to the index test according to the modified Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool. The meta-analysis on differential diagnosis of autoimmune pancreatitis versus pancreatic cancer by CT and mass-forming pancreatitis versus pancreatic cancer by MRI showed diagnostic odds ratios (95% confidence intervals) of, respectively, 189.63 (79.65–451.48) and 135.70 (36.17–509.13), both rated as weak evidence mainly due to the insufficient sample size. Conclusions More research on prognosis of acute pancreatitis is encouraged. The current pancreatitis radiomics studies have insufficient quality and share common scientific disadvantages. The evidence level rating is feasible and necessary for bringing the field of radiomics from preclinical research area to clinical stage. Supplementary Information The online version contains supplementary material available at 10.1186/s13244-022-01279-4.
Collapse
|
24
|
de la Pinta C. Radiomics in pancreatic cancer for oncologist: Present and future. Hepatobiliary Pancreat Dis Int 2022; 21:356-361. [PMID: 34961674 DOI: 10.1016/j.hbpd.2021.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/07/2021] [Indexed: 02/05/2023]
Abstract
Radiomics is changing the world of medicine and more specifically the world of oncology. Early diagnosis and treatment improve the prognosis of patients with cancer. After treatment, the evaluation of the response will determine future treatments. In oncology, every change in treatment means a loss of therapeutic options and this is key in pancreatic cancer. Radiomics has been developed in oncology in the early diagnosis and differential diagnosis of benign and malignant lesions, in the evaluation of response, in the prediction of possible side effects, marking the risk of recurrence, survival and prognosis of the disease. Some studies have validated its use to differentiate normal tissues from tumor tissues with high sensitivity and specificity, and to differentiate cystic lesions and pancreatic neuroendocrine tumor grades with texture parameters. In addition, these parameters have been related to survival in patients with pancreatic cancer and to response to radiotherapy and chemotherapy. This review aimed to establish the current status of the use of radiomics in pancreatic cancer and future perspectives.
Collapse
Affiliation(s)
- Carolina de la Pinta
- Radiation Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain.
| |
Collapse
|
25
|
Yan G, Yan G, Li H, Liang H, Peng C, Bhetuwal A, McClure MA, Li Y, Yang G, Li Y, Zhao L, Fan X. Radiomics and Its Applications and Progress in Pancreatitis: A Current State of the Art Review. Front Med (Lausanne) 2022; 9:922299. [PMID: 35814756 PMCID: PMC9259974 DOI: 10.3389/fmed.2022.922299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/31/2022] [Indexed: 11/25/2022] Open
Abstract
Radiomics involves high-throughput extraction and analysis of quantitative information from medical images. Since it was proposed in 2012, there are some publications on the application of radiomics for (1) predicting recurrent acute pancreatitis (RAP), clinical severity of acute pancreatitis (AP), and extrapancreatic necrosis in AP; (2) differentiating mass-forming chronic pancreatitis (MFCP) from pancreatic ductal adenocarcinoma (PDAC), focal autoimmune pancreatitis (AIP) from PDAC, and functional abdominal pain (functional gastrointestinal diseases) from RAP and chronic pancreatitis (CP); and (3) identifying CP and normal pancreas, and CP risk factors and complications. In this review, we aim to systematically summarize the applications and progress of radiomics in pancreatitis and it associated situations, so as to provide reference for related research.
Collapse
Affiliation(s)
- Gaowu Yan
- Department of Radiology, Suining Central Hospital, Suining, China
| | - Gaowen Yan
- Department of Radiology, The First Hospital of Suining, Suining, China
| | - Hongwei Li
- Department of Radiology, The Third Hospital of Mianyang and Sichuan Mental Health Center, Mianyang, China
| | - Hongwei Liang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chen Peng
- Department of Gastroenterology, The First Hospital of Suining, Suining, China
| | - Anup Bhetuwal
- Sichuan Key Laboratory of Medical Imaging, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Morgan A. McClure
- Department of Radiology and Imaging, Institute of Rehabilitation and Development of Brain Function, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, China
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Yongmei Li
| | - Guoqing Yang
- Department of Radiology, Suining Central Hospital, Suining, China
- Guoqing Yang
| | - Yong Li
- Department of Radiology, Suining Central Hospital, Suining, China
- Yong Li
| | - Linwei Zhao
- Department of Radiology, Suining Central Hospital, Suining, China
| | - Xiaoping Fan
- Department of Radiology, Suining Central Hospital, Suining, China
| |
Collapse
|
26
|
Repeatability and Image Quality of IDEAL-IQ in Human Lumbar Vertebrae for Fat and Iron Quantification across Acquisition Parameters. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2229160. [PMID: 35720027 PMCID: PMC9203175 DOI: 10.1155/2022/2229160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/09/2022] [Accepted: 05/14/2022] [Indexed: 11/17/2022]
Abstract
Echo asymmetry and least square estimation-IQ (IDEAL-IQ) were used to quantify fat and iron to verify the effects of collection parameters on repeatability and image quality of water and fat in human vertebral body. Six IDEAL-IQ sequences were used to scan 48 healthy adult women. Reproducibility of fat and iron quantification and image quality were assessed for six IDEAL-IQ sequences. The results showed that the correlation index (0.987, 0.721) of FF and R2∗ between scans of sequence 2 was higher than that of other sequences, and the consistency of fat quantification was better than that of iron (0.860 vs. 0.579) (P < 0.001). Sequence 2 had the highest image quality score (4.9) and the lowest CV score (9.2%). In the FF figure, SNR (18.8) and CNR (17.8 ± 6.4) were the highest, while CV was the lowest (36.7%, 36.1%). In the R2∗ figure, sequence 3 had the highest SNR (21.8) and CNR (20.5), but its CV (51.8% and 56.1%) was significantly higher than that of sequence 2. The occurrence of fat-water exchange (FWS) was lowest in sequence 2 and sequence 4 (0, N = 96). In conclusion, the fat quantification of IDEAL-IQ was robust to the changes of collection parameters, and section thickness (ST) had a certain effect on maintaining good repeatability of R2∗ quantification. The higher the ST was, the better the image quality of FF and R2∗ was maintained and stable and the less the occurrence of FWS.
Collapse
|
27
|
Abstract
The basic pancreatic lesions include location, size, shape, number, capsule, calcification/calculi, hemorrhage, cystic degeneration, fibrosis, pancreatic duct alterations, and microvessel. One or more basic lesions form a kind of pancreatic disease. As recognizing the characteristic imaging features of pancreatic basic lesions and their relationships with pathology aids in differentiating the variety of pancreatic diseases. The purpose of this study is to review the pathological and imaging features of the basic pancreatic lesions.
Collapse
|
28
|
Pantelis AG, Panagopoulou PA, Lapatsanis DP. Artificial Intelligence and Machine Learning in the Diagnosis and Management of Gastroenteropancreatic Neuroendocrine Neoplasms—A Scoping Review. Diagnostics (Basel) 2022; 12:diagnostics12040874. [PMID: 35453922 PMCID: PMC9027316 DOI: 10.3390/diagnostics12040874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/21/2022] Open
Abstract
Neuroendocrine neoplasms (NENs) and tumors (NETs) are rare neoplasms that may affect any part of the gastrointestinal system. In this scoping review, we attempt to map existing evidence on the role of artificial intelligence, machine learning and deep learning in the diagnosis and management of NENs of the gastrointestinal system. After implementation of inclusion and exclusion criteria, we retrieved 44 studies with 53 outcome analyses. We then classified the papers according to the type of studied NET (26 Pan-NETs, 59.1%; 3 metastatic liver NETs (6.8%), 2 small intestinal NETs, 4.5%; colorectal, rectal, non-specified gastroenteropancreatic and non-specified gastrointestinal NETs had from 1 study each, 2.3%). The most frequently used AI algorithms were Supporting Vector Classification/Machine (14 analyses, 29.8%), Convolutional Neural Network and Random Forest (10 analyses each, 21.3%), Random Forest (9 analyses, 19.1%), Logistic Regression (8 analyses, 17.0%), and Decision Tree (6 analyses, 12.8%). There was high heterogeneity on the description of the prediction model, structure of datasets, and performance metrics, whereas the majority of studies did not report any external validation set. Future studies should aim at incorporating a uniform structure in accordance with existing guidelines for purposes of reproducibility and research quality, which are prerequisites for integration into clinical practice.
Collapse
Affiliation(s)
- Athanasios G. Pantelis
- 4th Department of Surgery, Evaggelismos General Hospital of Athens, 10676 Athens, Greece;
- Correspondence:
| | | | - Dimitris P. Lapatsanis
- 4th Department of Surgery, Evaggelismos General Hospital of Athens, 10676 Athens, Greece;
| |
Collapse
|
29
|
Yoshikawa T, Takenaka D, Ohno Y. Editorial for "MRI Radiomics-Based Nomogram From Primary Tumor for Pretreatment Prediction of Peripancreatic Lymph Node Metastasis in Pancreatic Ductal Adenocarcinoma: A Multicenter Study". J Magn Reson Imaging 2022; 55:840-841. [PMID: 35104005 DOI: 10.1002/jmri.28090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 12/17/2023] Open
Affiliation(s)
- Takeshi Yoshikawa
- Department of Diagnostic and Interventional Radiology, Hyogo Cancer Center, Akashi, Japan
| | - Daisuke Takenaka
- Department of Diagnostic and Interventional Radiology, Hyogo Cancer Center, Akashi, Japan
| | - Yoshiharu Ohno
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
30
|
Shi Z, Ma C, Huang X, Cao D. Magnetic Resonance Imaging Radiomics-Based Nomogram From Primary Tumor for Pretreatment Prediction of Peripancreatic Lymph Node Metastasis in Pancreatic Ductal Adenocarcinoma: A Multicenter Study. J Magn Reson Imaging 2022; 55:823-839. [PMID: 34997795 DOI: 10.1002/jmri.28048] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Determining the absence or presence of peripancreatic lymph nodal metastasis (PLNM) is important to the pathologic staging, prognostication, and guidance of treatment in pancreatic ductal adenocarcinoma (PDAC) patients. Computed tomography and MRI had a poor sensitivity and diagnostic accuracy in the assessment of PLNM. PURPOSES To develop and validate a 3 T MRI primary tumor radiomics-based nomogram from multicenter datasets for pretreatment prediction of the PLNM in PDAC patients. STUDY TYPE Retrospective. SUBJECTS A total of 251 patients (156 men and 95 women; mean age, 60.85 ± 8.23 years) with histologically confirmed pancreatic ductal adenocarcinoma from three hospitals. FIELD STRENGTH AND SEQUENCES A 3.0 T and fat-suppressed T1-weighted imaging. ASSESSMENT Quantitative imaging features were extracted from fat-suppressed T1-weighted (FS T1WI) images at the arterial phase. STATISTICAL TESTS Normally distributed data were compared by using t-tests, while the Mann-Whitney U test was used to evaluate non-normally distributed data. The diagnostic performances of the preoperative and postoperative nomograms were assessed in the external validation cohort with the area under receiver operating characteristics curve (AUC), calibration curve, and decision curve analysis (DCA). AUCs were compared with the De Long test. A p value below 0.05 was considered to be statistically significant. RESULTS The AUCs of magnetic resonance imaging (MRI) Rad-score were 0.868 (95% confidence level [CI]: 0.613-0.852) and 0.772 (95% CI: 0.659-0.879) in the training and internal validation cohort, respectively. The preoperative and postoperative nomograms could accurately predict PLNM in the training cohort (AUC = 0.909 and 0.851) and were validated in both the internal and external cohorts (AUC = 0.835 and 0.805, 0.808 and 0.733, respectively). DCA indicated that the two novel nomograms are of similar clinical usefulness. DATA CONCLUSION Pre-/postoperative nomograms and the constructed radiomics signature from primary tumor based on FS T1WI of arterial phase could serve as a potential tool to predict PLNM in patients with PDAC. EVIDENCE LEVEL: 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Zhenshan Shi
- Department of Radiology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350005, China
| | - Chengle Ma
- Department of Radiology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350005, China
| | - Xinming Huang
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350005, China
| | - Dairong Cao
- Department of Radiology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350005, China
| |
Collapse
|
31
|
Hirata K, Sugimori H, Fujima N, Toyonaga T, Kudo K. Artificial intelligence for nuclear medicine in oncology. Ann Nucl Med 2022; 36:123-132. [PMID: 35028877 DOI: 10.1007/s12149-021-01693-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 11/07/2021] [Indexed: 12/12/2022]
Abstract
As in all other medical fields, artificial intelligence (AI) is increasingly being used in nuclear medicine for oncology. There are many articles that discuss AI from the viewpoint of nuclear medicine, but few focus on nuclear medicine from the viewpoint of AI. Nuclear medicine images are characterized by their low spatial resolution and high quantitativeness. It is noted that AI has been used since before the emergence of deep learning. AI can be divided into three categories by its purpose: (1) assisted interpretation, i.e., computer-aided detection (CADe) or computer-aided diagnosis (CADx). (2) Additional insight, i.e., AI provides information beyond the radiologist's eye, such as predicting genes and prognosis from images. It is also related to the field called radiomics/radiogenomics. (3) Augmented image, i.e., image generation tasks. To apply AI to practical use, harmonization between facilities and the possibility of black box explanations need to be resolved.
Collapse
Affiliation(s)
- Kenji Hirata
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan. .,Department of Nuclear Medicine, Hokkaido University Hospital, Sapporo, Japan. .,Division of Medical AI Education and Research, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| | | | - Noriyuki Fujima
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan.,Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Kohsuke Kudo
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan.,Division of Medical AI Education and Research, Hokkaido University Graduate School of Medicine, Sapporo, Japan.,Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan.,Global Center for Biomedical Science and Engineering, Hokkaido University Faculty of Medicine, Sapporo, Japan
| |
Collapse
|
32
|
Casà C, Piras A, D’Aviero A, Preziosi F, Mariani S, Cusumano D, Romano A, Boskoski I, Lenkowicz J, Dinapoli N, Cellini F, Gambacorta MA, Valentini V, Mattiucci GC, Boldrini L. The impact of radiomics in diagnosis and staging of pancreatic cancer. Ther Adv Gastrointest Endosc 2022; 15:26317745221081596. [PMID: 35342883 PMCID: PMC8943316 DOI: 10.1177/26317745221081596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 02/02/2022] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Pancreatic cancer (PC) is one of the most aggressive tumours, and better risk stratification among patients is required to provide tailored treatment. The meaning of radiomics and texture analysis as predictive techniques are not already systematically assessed. The aim of this study is to assess the role of radiomics in PC. METHODS A PubMed/MEDLINE and Embase systematic review was conducted to assess the role of radiomics in PC. The search strategy was 'radiomics [All Fields] AND ("pancreas" [MeSH Terms] OR "pancreas" [All Fields] OR "pancreatic" [All Fields])' and only original articles referred to PC in humans in the English language were considered. RESULTS A total of 123 studies and 183 studies were obtained using the mentioned search strategy on PubMed and Embase, respectively. After the complete selection process, a total of 56 papers were considered eligible for the analysis of the results. Radiomics methods were applied in PC for assessment technical feasibility and reproducibility aspects analysis, risk stratification, biologic or genomic status prediction and treatment response prediction. DISCUSSION Radiomics seems to be a promising approach to evaluate PC from diagnosis to treatment response prediction. Further and larger studies are required to confirm the role and allowed to include radiomics parameter in a comprehensive decision support system.
Collapse
Affiliation(s)
- Calogero Casà
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | | | - Francesco Preziosi
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Silvia Mariani
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Davide Cusumano
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Angela Romano
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ivo Boskoski
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Rome, Italy
| | - Jacopo Lenkowicz
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Nicola Dinapoli
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesco Cellini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Antonietta Gambacorta
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Vincenzo Valentini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gian Carlo Mattiucci
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Boldrini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
33
|
Li W, Xu C, Ye Z. Prediction of Pancreatic Neuroendocrine Tumor Grading Risk Based on Quantitative Radiomic Analysis of MR. Front Oncol 2021; 11:758062. [PMID: 34868970 PMCID: PMC8637752 DOI: 10.3389/fonc.2021.758062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/26/2021] [Indexed: 11/13/2022] Open
Abstract
Background Pancreatic neuroendocrine tumors (PNETs) grade is very important for treatment strategy of PNETs. The present study aimed to find the quantitative radiomic features for predicting grades of PNETs in MR images. Materials and Methods Totally 48 patients but 51 lesions with a pathological tumor grade were subdivided into low grade (G1) group and intermediate grade (G2) group. The ROI was manually segmented slice by slice in 3D-T1 weighted sequence with and without enhancement. Statistical differences of radiomic features between G1 and G2 groups were analyzed using the independent sample t-test. Logistic regression analysis was conducted to find better predictors in distinguishing G1 and G2 groups. Finally, receiver operating characteristic (ROC) was constructed to assess diagnostic performance of each model. Results No significant difference between G1 and G2 groups (P > 0.05) in non-enhanced 3D-T1 images was found. Significant differences in the arterial phase analysis between the G1 and the G2 groups appeared as follows: the maximum intensity feature (P = 0.021); the range feature (P = 0.039). Multiple logistic regression analysis based on univariable model showed the maximum intensity feature (P=0.023, OR = 0.621, 95% CI: 0.433-0.858) was an independent predictor of G1 compared with G2 group, and the area under the curve (AUC) was 0.695. Conclusions The maximum intensity feature of radiomic features in MR images can help to predict PNETs grade risk.
Collapse
Affiliation(s)
- Wei Li
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chao Xu
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
34
|
Do RKG, Kambadakone A. Radiomics for CT Assessment of Vascular Contact in Pancreatic Adenocarcinoma. Radiology 2021; 301:623-624. [PMID: 34491133 PMCID: PMC8630530 DOI: 10.1148/radiol.2021211635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Richard K. G. Do
- From the Department of Radiology, Memorial Sloan-Kettering Cancer
Center, 1275 York Ave, H-710, New York, NY 10065 (R.K.G.D.); and Division of
Abdominal Imaging and Intervention, Massachusetts General Hospital, Boston, Mass
(A.K.)
| | - Avinash Kambadakone
- From the Department of Radiology, Memorial Sloan-Kettering Cancer
Center, 1275 York Ave, H-710, New York, NY 10065 (R.K.G.D.); and Division of
Abdominal Imaging and Intervention, Massachusetts General Hospital, Boston, Mass
(A.K.)
| |
Collapse
|
35
|
Satake H, Ishigaki S, Ito R, Naganawa S. Radiomics in breast MRI: current progress toward clinical application in the era of artificial intelligence. Radiol Med 2021; 127:39-56. [PMID: 34704213 DOI: 10.1007/s11547-021-01423-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/14/2021] [Indexed: 12/11/2022]
Abstract
Breast magnetic resonance imaging (MRI) is the most sensitive imaging modality for breast cancer diagnosis and is widely used clinically. Dynamic contrast-enhanced MRI is the basis for breast MRI, but ultrafast images, T2-weighted images, and diffusion-weighted images are also taken to improve the characteristics of the lesion. Such multiparametric MRI with numerous morphological and functional data poses new challenges to radiologists, and thus, new tools for reliable, reproducible, and high-volume quantitative assessments are warranted. In this context, radiomics, which is an emerging field of research involving the conversion of digital medical images into mineable data for clinical decision-making and outcome prediction, has been gaining ground in oncology. Recent development in artificial intelligence has promoted radiomics studies in various fields including breast cancer treatment and numerous studies have been conducted. However, radiomics has shown a translational gap in clinical practice, and many issues remain to be solved. In this review, we will outline the steps of radiomics workflow and investigate clinical application of radiomics focusing on breast MRI based on published literature, as well as current discussion about limitations and challenges in radiomics.
Collapse
Affiliation(s)
- Hiroko Satake
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| | - Satoko Ishigaki
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Rintaro Ito
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| |
Collapse
|
36
|
Neuroendocrine Carcinomas of the Digestive Tract: What Is New? Cancers (Basel) 2021; 13:cancers13153766. [PMID: 34359666 PMCID: PMC8345167 DOI: 10.3390/cancers13153766] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroendocrine carcinomas (NEC) are rare tumors with a rising incidence. They show poorly differentiated morphology with a high proliferation rate (Ki-67 index). They frequently arise in the lung (small and large-cell lung cancer) but rarely from the gastrointestinal tract. Due to their rarity, very little is known about digestive NEC and few studies have been conducted. Therefore, most of therapeutic recommendations are issued from work on small-cell lung cancers (SCLC). Recent improvement in pathology and imaging has allowed for better detection and classification of high-grade NEN. The 2019 World Health Organization (WHO) classification has described a new entity of well-differentiated grade 3 neuroendocrine tumors (NET G-3), with better prognosis, that should be managed separately from NEC. NEC are aggressive neoplasms often diagnosed at a metastatic state. In the localized setting, surgery can be performed in selected patients followed by adjuvant platinum-based chemotherapy. Concurrent chemoradiotherapy is also an option for NEC of the lung, rectum, and esophagus. In metastatic NEC, chemotherapy is administered with a classic combination of platinum salts and etoposide in the first-line setting. Peptide receptor radionuclide therapy (PRRT) has shown positive results in high-grade NEN populations and immunotherapy trials are still ongoing. Available therapies have improved the overall survival of NEC but there is still an urgent need for improvement. This narrative review sums up the current data on digestive NEC while exploring future directions for their management.
Collapse
|
37
|
Pancreatic cancer epidemiology: understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol 2021; 18:493-502. [PMID: 34002083 PMCID: PMC9265847 DOI: 10.1038/s41575-021-00457-x] [Citation(s) in RCA: 521] [Impact Index Per Article: 130.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/20/2021] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer is a leading cause of cancer death worldwide and its global burden has more than doubled over the past 25 years. The highest incidence regions for pancreatic cancer include North America, Europe and Australia, and although much of this increase is due to ageing worldwide populations, there are key modifiable risk factors for pancreatic cancer such as cigarette smoking, obesity, diabetes and alcohol intake. The prevalence of these risk factors is increasing in many global regions, resulting in increasing age-adjusted incidence rates for pancreatic cancer, but the relative contribution from these risk factors varies globally due to variation in the underlying prevalence and prevention strategies. Inherited genetic factors, although not directly modifiable, are an important component of pancreatic cancer risk, and include pathogenic variants in hereditary cancer genes, genes associated with hereditary pancreatitis, as well as common variants identified in genome-wide association studies. Identification of the genetic changes that underlie pancreatic cancer not only provides insight into the aetiology of this cancer but also provides an opportunity to guide early detection strategies. The goal of this Review is to provide an up-to-date overview of the established modifiable and inherited risk factors for pancreatic cancer.
Collapse
|
38
|
Barat M, Chassagnon G, Dohan A, Gaujoux S, Coriat R, Hoeffel C, Cassinotto C, Soyer P. Artificial intelligence: a critical review of current applications in pancreatic imaging. Jpn J Radiol 2021; 39:514-523. [PMID: 33550513 DOI: 10.1007/s11604-021-01098-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022]
Abstract
The applications of artificial intelligence (AI), including machine learning and deep learning, in the field of pancreatic disease imaging are rapidly expanding. AI can be used for the detection of pancreatic ductal adenocarcinoma and other pancreatic tumors but also for pancreatic lesion characterization. In this review, the basic of radiomics, recent developments and current results of AI in the field of pancreatic tumors are presented. Limitations and future perspectives of AI are discussed.
Collapse
Affiliation(s)
- Maxime Barat
- Department of Radiology, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 27 Rue du Faubourg Saint-Jacques, Paris, France
- Université de Paris, Descartes-Paris 5, 75006, Paris, France
| | - Guillaume Chassagnon
- Department of Radiology, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 27 Rue du Faubourg Saint-Jacques, Paris, France
- Université de Paris, Descartes-Paris 5, 75006, Paris, France
| | - Anthony Dohan
- Department of Radiology, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 27 Rue du Faubourg Saint-Jacques, Paris, France
- Université de Paris, Descartes-Paris 5, 75006, Paris, France
| | - Sébastien Gaujoux
- Université de Paris, Descartes-Paris 5, 75006, Paris, France
- Department of Abdominal Surgery, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 75014, Paris, France
| | - Romain Coriat
- Université de Paris, Descartes-Paris 5, 75006, Paris, France
- Department of Gastroenterology, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 75014, Paris, France
| | - Christine Hoeffel
- Department of Radiology, Robert Debré Hospital, 51092, Reims, France
| | - Christophe Cassinotto
- Department of Radiology, CHU Montpellier, University of Montpellier, Saint-Éloi Hospital, 34000, Montpellier, France
| | - Philippe Soyer
- Department of Radiology, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 27 Rue du Faubourg Saint-Jacques, Paris, France.
- Université de Paris, Descartes-Paris 5, 75006, Paris, France.
| |
Collapse
|
39
|
Pellat A, Cottereau AS, Palmieri LJ, Soyer P, Marchese U, Brezault C, Coriat R. Digestive Well-Differentiated Grade 3 Neuroendocrine Tumors: Current Management and Future Directions. Cancers (Basel) 2021; 13:2448. [PMID: 34070035 PMCID: PMC8158108 DOI: 10.3390/cancers13102448] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 02/06/2023] Open
Abstract
Digestive well-differentiated grade 3 neuroendocrine tumors (NET G-3) have been clearly defined since the 2017 World Health Organization classification. They are still a rare category lacking specific data and standardized management. Their distinction from other types of neuroendocrine neoplasms (NEN) not only lies in morphology but also in genotype, aggressiveness, functional imaging uptake, and treatment response. Most of the available data comes from pancreatic series, which is the most frequent tumor site for this entity. In the non-metastatic setting, surgical resection is recommended, irrespective of grade and tumor site. For metastatic NET G-3, chemotherapy is the main first-line treatment with temozolomide-based regimen showing more efficacy than platinum-based regimen, especially when Ki-67 index <55%. Targeted therapies, such as sunitinib and everolimus, have also shown some positive therapeutic efficacy in small samples of patients. Functional imaging plays a key role for detection but also treatment selection. In the second or further-line setting, peptide receptor radionuclide therapy has shown promising response rates in high-grade NEN. Finally, immunotherapy is currently investigated as a new therapeutic approach with trials still ongoing. More data will come with future work now focusing on this specific subgroup. The aim of this review is to summarize the current data on digestive NET G-3 and explore future directions for their management.
Collapse
Affiliation(s)
- Anna Pellat
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, AP-HP, 27 rue du Faubourg Saint Jacques, Université de Paris, 75014 Paris, France; (L.-J.P.); (C.B.); (R.C.)
| | - Anne Ségolène Cottereau
- Department of Nuclear Medicine, Hôpital Cochin, AP-HP, 27 rue du Faubourg Saint Jacques, Université de Paris, 75014 Paris, France;
| | - Lola-Jade Palmieri
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, AP-HP, 27 rue du Faubourg Saint Jacques, Université de Paris, 75014 Paris, France; (L.-J.P.); (C.B.); (R.C.)
| | - Philippe Soyer
- Department of Radiology, Hôpital Cochin, AP-HP, 27 rue du Faubourg Saint Jacques, Université de Paris, 75014 Paris, France;
| | - Ugo Marchese
- Department of Surgery, Hôpital Cochin, AP-HP, 27 rue du Faubourg Saint Jacques, Université de Paris, 75014 Paris, France;
| | - Catherine Brezault
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, AP-HP, 27 rue du Faubourg Saint Jacques, Université de Paris, 75014 Paris, France; (L.-J.P.); (C.B.); (R.C.)
| | - Romain Coriat
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, AP-HP, 27 rue du Faubourg Saint Jacques, Université de Paris, 75014 Paris, France; (L.-J.P.); (C.B.); (R.C.)
| |
Collapse
|
40
|
Barat M, Hoeffel C, Aissaoui M, Dohan A, Oudjit A, Dautry R, Paisant A, Malgras B, Cottereau AS, Soyer P. Focal splenic lesions: Imaging spectrum of diseases on CT, MRI and PET/CT. Diagn Interv Imaging 2021; 102:501-513. [PMID: 33965354 DOI: 10.1016/j.diii.2021.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022]
Abstract
The spleen can be affected by a variety of diseases. Some of them are readily identified as variations of normal or benign diseases on imaging. However, for a substantial number of focal splenic abnormalities, the diagnosis can be difficult so that histopathologic analysis may be required for a definite diagnosis. In this review, the typical splenic abnormalities that can be diagnosed with imaging with a high degree of confidence are illustrated. The complementary role of computed tomography (CT), magnetic resonance imaging and positron emission tomography/CT that helps make a diagnostic approach is discussed. Finally, current applications and future trends of radiomics and artificial intelligence for the diagnosis of splenic diseases are addressed.
Collapse
Affiliation(s)
- Maxime Barat
- Department of Radiology, Hôpital Cochin, AP-HP, 75014 Paris, France; Université de Paris, 75006 Paris, France.
| | - Christine Hoeffel
- Department of Radiology, Reims University Hospital, 51092 Reims, France; CRESTIC, University of Reims Champagne-Ardenne, 51100 Reims, France
| | | | - Anthony Dohan
- Department of Radiology, Hôpital Cochin, AP-HP, 75014 Paris, France; Université de Paris, 75006 Paris, France
| | - Amar Oudjit
- Department of Radiology, Hôpital Cochin, AP-HP, 75014 Paris, France
| | - Raphael Dautry
- Department of Radiology, Hôpital Cochin, AP-HP, 75014 Paris, France
| | - Anita Paisant
- Department of Radiology, University Hospital of Angers, 49100 Angers, France; Faculté de Médecine, Université d'Angers, 49045 Angers, France
| | - Brice Malgras
- Department of Digestive and Endocrine Surgery, Bégin Army Training hospital, 94160 Saint-Mandé, France; École du Val-de-Grâce, 75005 Paris, France
| | - Anne-Ségolène Cottereau
- Université de Paris, 75006 Paris, France; Department of Nuclear Medicine, Hôpital Cochin, AP-HP, 75014 Paris, France
| | - Philippe Soyer
- Department of Radiology, Hôpital Cochin, AP-HP, 75014 Paris, France; Université de Paris, 75006 Paris, France
| |
Collapse
|
41
|
Liang L, Ding Y, Yu Y, Liu K, Rao S, Ge Y, Zeng M. Whole-tumour evaluation with MRI and radiomics features to predict the efficacy of S-1 for adjuvant chemotherapy in postoperative pancreatic cancer patients: a pilot study. BMC Med Imaging 2021; 21:75. [PMID: 33902469 PMCID: PMC8077911 DOI: 10.1186/s12880-021-00605-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 04/14/2021] [Indexed: 01/13/2023] Open
Abstract
Background Multiple guidelines for pancreatic ductal adenocarcinoma (PDAC) suggest that all stages of patients need to receive postoperative adjuvant chemotherapy. S-1 is a recently emerged oral antitumour agent recommended by the guidelines. However, which population would benefit from S-1 needs to be determined, and predictors of chemotherapy response are needed for personalized precision medicine. This pilot study aimed to initially identify whether whole-tumour evaluation with MRI and radiomics features could be used for predicting the efficacy of S-1 and to find potential predictors of the efficacy of S-1 as evidence to assist personalized precision treatment. Methods Forty-six patients with PDAC (31 in the primary cohort and 15 in the validation cohort) who underwent curative resection and subsequently adjuvant chemotherapy with S-1 were included. Pre-operative abdominal contrast-enhanced MRI was performed, and radiomics features of the whole PDAC were extracted from the primary cohort. After univariable analysis and radiomics features selection, a multivariable Cox regression model for survival analysis was subsequently used to select statistically significant factors associated with postoperative disease-free survival (DFS). Predictive capacities of the factors were tested on the validation cohort by using Kaplan–Meier method. Results Multivariable Cox regression analysis identified the probability of T1WI_NGTDM_Strength and tumour location as independent predictors of the efficacy of S-1 for adjuvant chemotherapy of PDAC (p = 0.005 and 0.013) in the primary cohort, with hazard ratios (HRs) of 0.289 and 0.293, respectively. Further survival analysis showed that patients in the low-T1WI_NGTDM_Strength group had shorter DFS (median = 5.1 m) than those in the high-T1WI_NGTDM_Strength group (median = 13.0 m) (p = 0.006), and patients with PDAC on the pancreatic head exhibited shorter DFS (median = 7.0 m) than patients with tumours in other locations (median = 20.0 m) (p = 0.016). In the validation cohort, the difference in DFS between patients with low-T1WI_NGTDM_Strength and high-T1WI_NGTDM_Strength and the difference between patients with PDAC on the pancreatic head and that in other locations were approved, with marginally significant (p = 0.073 and 0.050), respectively. Conclusions Whole-tumour radiomics feature of T1WI_NGTDM_Strength and tumour location were potential predictors of the efficacy of S-1 and for the precision selection of S-1 as adjuvant chemotherapy regimen for PDAC.
Collapse
Affiliation(s)
- Liang Liang
- Department of Radiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Ying Ding
- Department of Radiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Kai Liu
- Department of Radiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Shengxiang Rao
- Department of Radiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Yingqian Ge
- Siemens Healthineers, No. 278 Zhou Zhu Road, Pudong New District, Shanghai, 201318, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, and Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
42
|
D’Onofrio M, De Robertis R, Aluffi G, Cadore C, Beleù A, Cardobi N, Malleo G, Manfrin E, Bassi C. CT Simplified Radiomic Approach to Assess the Metastatic Ductal Adenocarcinoma of the Pancreas. Cancers (Basel) 2021; 13:cancers13081843. [PMID: 33924363 PMCID: PMC8069159 DOI: 10.3390/cancers13081843] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to perform a simplified radiomic analysis of pancreatic ductal adenocarcinoma based on qualitative and quantitative tumor features and to compare the results between metastatic and non-metastatic patients. A search of our radiological, surgical, and pathological databases identified 1218 patients with a newly diagnosed pancreatic ductal adenocarcinoma who were referred to our Institution between January 2014 and December 2018. Computed Tomography (CT) examinations were reviewed analyzing qualitative and quantitative features. Two hundred eighty-eight patients fulfilled the inclusion criteria and were included in this study. Overall, metastases were present at diagnosis in 86/288 patients, while no metastases were identified in 202/288 patients. Ill-defined margins and a hypodense appearance on portal-phase images were significantly more common among patients with metastases compared to non-metastatic patients (p < 0.05). Metastatic tumors showed a significantly larger size and significantly lower arterial index, perfusion index, and permeability index compared to non-metastatic tumors (p < 0.05). In the management of pancreatic ductal adenocarcinoma, early detection and correct staging are key elements. The study of computerized tomography characteristics of pancreatic ductal adenocarcinoma showed substantial differences, both qualitative and quantitative, between metastatic and non-metastatic disease.
Collapse
Affiliation(s)
- Mirko D’Onofrio
- Department of Radiology, G.B. Rossi Hospital, University of Verona, 37129 Verona, Italy; (G.A.); (C.C.); (A.B.)
- Correspondence:
| | - Riccardo De Robertis
- Department of Radiology, Ospedale Civile Maggiore, Azienda Ospedaliera Universitaria Integrata Verona, 37126 Verona, Italy; (R.D.R.); (N.C.)
| | - Gregorio Aluffi
- Department of Radiology, G.B. Rossi Hospital, University of Verona, 37129 Verona, Italy; (G.A.); (C.C.); (A.B.)
| | - Camilla Cadore
- Department of Radiology, G.B. Rossi Hospital, University of Verona, 37129 Verona, Italy; (G.A.); (C.C.); (A.B.)
| | - Alessandro Beleù
- Department of Radiology, G.B. Rossi Hospital, University of Verona, 37129 Verona, Italy; (G.A.); (C.C.); (A.B.)
| | - Nicolò Cardobi
- Department of Radiology, Ospedale Civile Maggiore, Azienda Ospedaliera Universitaria Integrata Verona, 37126 Verona, Italy; (R.D.R.); (N.C.)
| | - Giuseppe Malleo
- Unit of General and Pancreatic Surgery, The Pancreas Institute, Policlinico GB Rossi, University of Verona, 37129 Verona, Italy; (G.M.); (C.B.)
| | - Erminia Manfrin
- Department of Pathology, G.B. Rossi Hospital, University of Verona, 37129 Verona, Italy;
| | - Claudio Bassi
- Unit of General and Pancreatic Surgery, The Pancreas Institute, Policlinico GB Rossi, University of Verona, 37129 Verona, Italy; (G.M.); (C.B.)
| |
Collapse
|
43
|
Hirata K, Tamaki N. Quantitative FDG PET Assessment for Oncology Therapy. Cancers (Basel) 2021; 13:cancers13040869. [PMID: 33669531 PMCID: PMC7922629 DOI: 10.3390/cancers13040869] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary PET enables quantitative assessment of tumour biology in vivo. Accumulation of F-18 fluorodeoxyglucose (FDG) may reflect tumour metabolic activity. Quantitative assessment of FDG uptake can be applied for treatment monitoring. Numerous studies indicated biochemical change assessed by FDG-PET as a more sensitive marker than morphological change. Those with complete metabolic response after therapy may show better prognosis. Assessment of metabolic change may be performed using absolute FDG uptake or metabolic tumour volume. More recently, radiomics approaches have been applied to FDG PET. Texture analysis quantifies intratumoral heterogeneity in a voxel-by-voxel basis. Combined with various machine learning techniques, these new quantitative parameters hold a promise for assessing tissue characterization and predicting treatment effect, and could also be used for future prognosis of various tumours. Abstract Positron emission tomography (PET) has unique characteristics for quantitative assessment of tumour biology in vivo. Accumulation of F-18 fluorodeoxyglucose (FDG) may reflect tumour characteristics based on its metabolic activity. Quantitative assessment of FDG uptake can often be applied for treatment monitoring after chemotherapy or chemoradiotherapy. Numerous studies indicated biochemical change assessed by FDG PET as a more sensitive marker than morphological change estimated by CT or MRI. In addition, those with complete metabolic response after therapy may show better disease-free survival and overall survival than those with other responses. Assessment of metabolic change may be performed using absolute FDG uptake in the tumour (standardized uptake value: SUV). In addition, volumetric parameters such as metabolic tumour volume (MTV) have been introduced for quantitative assessment of FDG uptake in tumour. More recently, radiomics approaches that focus on image-based precision medicine have been applied to FDG PET, as well as other radiological imaging. Among these, texture analysis extracts intratumoral heterogeneity on a voxel-by-voxel basis. Combined with various machine learning techniques, these new quantitative parameters hold a promise for assessing tissue characterization and predicting treatment effect, and could also be used for future prognosis of various tumours, although multicentre clinical trials are needed before application in clinical settings.
Collapse
Affiliation(s)
- Kenji Hirata
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan;
| | - Nagara Tamaki
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Correspondence:
| |
Collapse
|