1
|
Barat M, Greffier J, Si-Mohamed S, Dohan A, Pellat A, Frandon J, Calame P, Soyer P. CT Imaging of the Pancreas: A Review of Current Developments and Applications. Can Assoc Radiol J 2025:8465371251319965. [PMID: 39985297 DOI: 10.1177/08465371251319965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2025] Open
Abstract
Pancreatic cancer continues to pose daily challenges to clinicians, radiologists, and researchers. These challenges are encountered at each stage of pancreatic cancer management, including early detection, definite characterization, accurate assessment of tumour burden, preoperative planning when surgical resection is possible, prediction of tumour aggressiveness, response to treatment, and detection of recurrence. CT imaging of the pancreas has made major advances in recent years through innovations in research and clinical practice. Technical advances in CT imaging, often in combination with imaging biomarkers, hold considerable promise in addressing such challenges. Ongoing research in dual-energy and spectral photon-counting computed tomography, new applications of artificial intelligence and image rendering have led to innovative implementations that allow now a more precise diagnosis of pancreatic cancer and other diseases affecting this organ. This article aims to explore the major research initiatives and technological advances that are shaping the landscape of CT imaging of the pancreas. By highlighting key contributions in diagnostic imaging, artificial intelligence, and image rendering, this article provides a comprehensive overview of how these innovations are enhancing diagnostic precision and improving outcome in patients with pancreatic diseases.
Collapse
Affiliation(s)
- Maxime Barat
- Université Paris Cité, Faculté de Médecine, Paris, Île-de-France, France
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France
| | - Joël Greffier
- Department of Medical Imaging, PRIM Platform, Nîmes University Hospital, University of Montpellier, Medical Imaging Group Nîmes, IMAGINE UR UM 103, Nîmes, France
| | - Salim Si-Mohamed
- University of Lyon, INSA-Lyon, University Claude Bernard Lyon 1, UJM-Saint Etienne, CNRS, Inserm, CREATIS UMR 5220, U1206, Villeurbanne, France
- Department of Radiology, Louis Pradel Hospital, Hospices Civils de Lyon, Bron, Auvergne-Rhône-Alpes, France
| | - Anthony Dohan
- Université Paris Cité, Faculté de Médecine, Paris, Île-de-France, France
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France
| | - Anna Pellat
- Université Paris Cité, Faculté de Médecine, Paris, Île-de-France, France
- Gastroenterology, Endoscopy and Digestive Oncology Unit, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France
| | - Julien Frandon
- Department of Medical Imaging, PRIM Platform, Nîmes University Hospital, University of Montpellier, Medical Imaging Group Nîmes, IMAGINE UR UM 103, Nîmes, France
| | - Paul Calame
- Department of Radiology, University of Franche-Comté, CHRU Besançon, Besançon, France
- EA 4662 Nanomedicine Lab, Imagery and Therapeutics, University of Franche-Comté, Besançon, Bourgogne-Franche-Comté, France
| | - Philippe Soyer
- Université Paris Cité, Faculté de Médecine, Paris, Île-de-France, France
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France
| |
Collapse
|
2
|
Belfrage H, Kuuliala K, Kuuliala A, Mustonen H, Puolakkainen P, Kylänpää L, Seppänen H, Louhimo J. Circulating necroptosis markers in chronic pancreatitis and pancreatic cancer: Associations with diagnosis and prognostic factors. Pancreatology 2024; 24:1229-1236. [PMID: 39613683 DOI: 10.1016/j.pan.2024.11.016] [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: 09/07/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
OBJECTIVES Necroptosis, a programmed inflammatory cell death, is implicated in the pathogenesis of pancreatitis and its potential progression to pancreatic ductal adenocarcinoma (PDAC), but its role remains unclear. We compared plasma levels of necroptosis-related markers - mixed lineage kinase domain-like protein (MLKL), interleukin (IL)-33, and its soluble receptor (sST2)- in patients with chronic pancreatitis (CP), PDAC, and healthy controls (HC), to explore their diagnostic and prognostic significance. METHODS Plasma was collected from patients pre-procedurally (PDAC, n = 82; CP, n = 25) and from HC (n = 39), and studied by ELISA. Clinical and routine laboratory data were collected, and pancreas was defined as soft or non-soft based on intraoperative or imaging findings. Mann-Whitney U test, Spearman correlation coefficients, ROC analysis were used for statistical analysis. RESULTS Plasma MLKL was lower in patients with CP than in other groups (p < 0.001) and PDAC patients treated with upfront surgery (PDACUS, n = 65) had lower MLKL than HC (p = 0.035). MLKL differentiated CP from PDACUS (AUC 0.83, p < 0.001). sST2 levels were significantly lower in HC than in other groups (p < 0.001) and in PDAC patients with a soft pancreas compared with non-soft (p < 0.005). In Lewis antigen positive PDACUS (n = 59), sST2 had positive correlations with CA19-9 measured concurrently and after 1 month, and with CEA measured after 1 or 6 months. CONCLUSIONS Circulating levels of MLKL are lower in patients with CP than PDAC. Elevated sST2 levels are associated with pancreatic diseases. Further studies are required to show whether MLKL and sST2 could be useful biomarkers in evaluating pancreatic diseases.
Collapse
Affiliation(s)
- Hanna Belfrage
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.
| | - Krista Kuuliala
- Department of Bacteriology and Immunology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Antti Kuuliala
- Department of Bacteriology and Immunology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Harri Mustonen
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Translational Cancer Medicine Program and ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Pauli Puolakkainen
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Translational Cancer Medicine Program and ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Leena Kylänpää
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Hanna Seppänen
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Translational Cancer Medicine Program and ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Johanna Louhimo
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
3
|
Bülbül HM, Burakgazi G, Kesimal U, Kaba E. Radiomics-based machine learning in the differentiation of benign and malignant bowel wall thickening radiomics in bowel wall thickening. Jpn J Radiol 2024; 42:872-879. [PMID: 38536559 DOI: 10.1007/s11604-024-01558-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/12/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE To distinguish malignant and benign bowel wall thickening (BWT) by using computed tomography (CT) texture features based on machine learning (ML) models and to compare its success with the clinical model and combined model. METHODS One hundred twenty-two patients with BWT identified on contrast-enhanced abdominal CT and underwent colonoscopy were included in this retrospective study. Texture features were extracted from CT images using LifeX software. Feature selection and reduction were performed using the Least Absolute Shrinkage and Selection Operator (LASSO). Six radiomic features were selected with LASSO. In the clinical model, six features (age, gender, thickness, fat stranding, symmetry, and lymph node) were included. Six radiomic and six clinical features were used in the combined model. Classification was done using two machine learning algorithms: Support Vector Machine (SVM) and Logistic Regression (LR). The data sets were divided into 80% training set and 20% test set. Then, training took place with all three datasets. The model's success was tested with the test set consisting of features not used during training. RESULTS In the training set, the combined model had the best performance with the area under the curve (AUC) value of 0.99 for SVM and 0.95 for LR. In the radiomic-derived model, the AUC value is 0.87 in SVM and 0.79 in LR. In the clinical model, SVM made this distinction with 0.95 AUC and LR with 0.92 AUC value. In the test set, the classifier with the highest success distinguishing malignant wall thickening is SVM in the radiomic-derived model with an AUC value of 0.90. In other models, the AUC value is in the range of 0.75-0.86, and the accuracy values are in the range of 0.72-0.84. CONCLUSION In conclusion, radiomic-based machine learning has shown high success in distinguishing malignant and benign BWT and may improve diagnostic accuracy compared to clinical features only. The results of our study may help ensure early diagnosis and treatment of colorectal cancers by facilitating the recognition of malignant BWT.
Collapse
Affiliation(s)
- Hande Melike Bülbül
- Department of Radiology, Ministry of Health Recep Tayyip Erdoğan University Training and Research Hospital, Rize, Turkey.
| | - Gülen Burakgazi
- Department of Radiology, Ministry of Health Recep Tayyip Erdoğan University Training and Research Hospital, Rize, Turkey
| | - Uğur Kesimal
- Department of Radiology, Ministry of Health Ankara Training and Research Hospital, Ankara, Turkey
| | - Esat Kaba
- Department of Radiology, Ministry of Health Recep Tayyip Erdoğan University Training and Research Hospital, Rize, Turkey
| |
Collapse
|
4
|
Cai TN, Zhao L, Yang Y, Mao HM, Huang SG, Guo WL. Development of a CT-based radiomics-clinical model to diagnose acute pancreatitis on nonobvious findings on CT in children with pancreaticobiliary maljunction. Br J Radiol 2024; 97:1029-1037. [PMID: 38460184 PMCID: PMC11075976 DOI: 10.1093/bjr/tqae054] [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: 10/14/2023] [Revised: 02/19/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
OBJECTIVES Since neither abdominal pain nor pancreatic enzyme elevation is specific for acute pancreatitis (AP), the diagnosis of AP in patients with pancreaticobiliary maljunction (PBM) may be challenging when the pancreas appears normal or nonobvious on CT. This study aimed to develop a quantitative radiomics-based nomogram of pancreatic CT for identifying AP in children with PBM who have nonobvious findings on CT. METHODS PBM patients with a diagnosis of AP evaluated at the Children's Hospital of Soochow University from June 2015 to October 2022 were retrospectively reviewed. The radiological features and clinical factors associated with AP were evaluated. Based on the selected variables, multivariate logistic regression was used to construct clinical, radiomics, and combined models. RESULTS Two clinical parameters and 6 radiomics characteristics were chosen based on their significant association with AP, as demonstrated in the training (area under curve [AUC]: 0.767, 0.892) and validation (AUC: 0.757, 0.836) datasets. The radiomics-clinical nomogram demonstrated superior performance in both the training (AUC, 0.938) and validation (AUC, 0.864) datasets, exhibiting satisfactory calibration (P > .05). CONCLUSIONS Our radiomics-based nomogram is an accurate, noninvasive diagnostic technique that can identify AP in children with PBM even when CT presentation is not obvious. ADVANCES IN KNOWLEDGE This study extracted imaging features of nonobvious pancreatitis. Then it developed and evaluated a combined model with these features.
Collapse
Affiliation(s)
- Tian-na Cai
- Department of Radiology, Children’s Hospital of Soochow University, Suzhou 215025, China
| | - Lian Zhao
- Department of Radiology, Children’s Hospital of Soochow University, Suzhou 215025, China
| | - Yang Yang
- Department of Radiology, Children’s Hospital of Soochow University, Suzhou 215025, China
| | - Hui-min Mao
- Department of Radiology, Children’s Hospital of Soochow University, Suzhou 215025, China
| | - Shun-gen Huang
- Pediatric Surgery, Children’s Hospital of Soochow University, Suzhou 215025, China
| | - Wan-liang Guo
- Department of Radiology, Children’s Hospital of Soochow University, Suzhou 215025, China
| |
Collapse
|
5
|
Yu C, Ruan Y, Yu L, Wang X, Hu Z, Zhu G, Huang T. Predicting postoperative prognosis of pancreatic cancer using a computed tomography-based radio-clinical model: exploring biologic functions. J Gastrointest Surg 2024; 28:458-466. [PMID: 38583896 DOI: 10.1016/j.gassur.2024.02.005] [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: 10/31/2023] [Revised: 01/19/2024] [Accepted: 02/03/2024] [Indexed: 04/09/2024]
Abstract
Computed tomography (CT) imaging has the potential to assist in predicting the prognosis and treatment strategies for pancreatic cancer (PC). This study aimed to develop and validate a radio-clinical model based on preoperative multiphase CT assessments to predict the overall survival (OS) of PC and identify differentially expressed genes associated with OS. METHODS Patients with PC who had undergone radical pancreatectomy (R0 resection) were divided into development and external validation sets. Independent predictors of OS were identified using Cox regression analyses and included in the nomogram, which was externally validated. The area under the curve was used to measure the model's accuracy in estimating OS probability. RNA sequencing data from The Cancer Genome Atlas were used for gene expression analysis. RESULTS In the development and external validation sets, survival was estimated respectively for 132 and 27 patients. Multivariate Cox regression analysis identified 5 independent OS predictors: age (P = .049), sex (P = .001), bilirubin level (P = .005), tumor size (P = .020), and venous invasion (P = .041). These variables were incorporated into the nomogram. Patients were divided into high- and low-risk groups for OS and survival curves showed that all patients in the low-risk group had better OS than that of those in the high-risk group (P < .001). Differentially expressed genes in patients with a poor prognosis were involved in neuroactive ligand-receptor interaction. CONCLUSION The radio-clinical model may be clinically useful for successfully predicting PC prognosis.
Collapse
Affiliation(s)
- Can Yu
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuli Ruan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lan Yu
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinxin Wang
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhaoshen Hu
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guanyu Zhu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tao Huang
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
6
|
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
|
7
|
Bette S, Canalini L, Feitelson LM, Woźnicki P, Risch F, Huber A, Decker JA, Tehlan K, Becker J, Wollny C, Scheurig-Münkler C, Wendler T, Schwarz F, Kroencke T. Radiomics-Based Machine Learning Model for Diagnosis of Acute Pancreatitis Using Computed Tomography. Diagnostics (Basel) 2024; 14:718. [PMID: 38611632 PMCID: PMC11011980 DOI: 10.3390/diagnostics14070718] [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: 02/02/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
In the early diagnostic workup of acute pancreatitis (AP), the role of contrast-enhanced CT is to establish the diagnosis in uncertain cases, assess severity, and detect potential complications like necrosis, fluid collections, bleeding or portal vein thrombosis. The value of texture analysis/radiomics of medical images has rapidly increased during the past decade, and the main focus has been on oncological imaging and tumor classification. Previous studies assessed the value of radiomics for differentiating between malignancies and inflammatory diseases of the pancreas as well as for prediction of AP severity. The aim of our study was to evaluate an automatic machine learning model for AP detection using radiomics analysis. Patients with abdominal pain and contrast-enhanced CT of the abdomen in an emergency setting were retrospectively included in this single-center study. The pancreas was automatically segmented using TotalSegmentator and radiomics features were extracted using PyRadiomics. We performed unsupervised hierarchical clustering and applied the random-forest based Boruta model to select the most important radiomics features. Important features and lipase levels were included in a logistic regression model with AP as the dependent variable. The model was established in a training cohort using fivefold cross-validation and applied to the test cohort (80/20 split). From a total of 1012 patients, 137 patients with AP and 138 patients without AP were included in the final study cohort. Feature selection confirmed 28 important features (mainly shape and first-order features) for the differentiation between AP and controls. The logistic regression model showed excellent diagnostic accuracy of radiomics features for the detection of AP, with an area under the curve (AUC) of 0.932. Using lipase levels only, an AUC of 0.946 was observed. Using both radiomics features and lipase levels, we showed an excellent AUC of 0.933 for the detection of AP. Automated segmentation of the pancreas and consecutive radiomics analysis almost achieved the high diagnostic accuracy of lipase levels, a well-established predictor of AP, and might be considered an additional diagnostic tool in unclear cases. This study provides scientific evidence that automated image analysis of the pancreas achieves comparable diagnostic accuracy to lipase levels and might therefore be used in the future in the rapidly growing era of AI-based image analysis.
Collapse
Affiliation(s)
- Stefanie Bette
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
| | - Luca Canalini
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
| | - Laura-Marie Feitelson
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
| | - Piotr Woźnicki
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, University of Würzburg, 97080 Würzburg, Germany;
| | - Franka Risch
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
| | - Adrian Huber
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
| | - Josua A. Decker
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
| | - Kartikay Tehlan
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
| | - Judith Becker
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
| | - Claudia Wollny
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
| | - Christian Scheurig-Münkler
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
| | - Thomas Wendler
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
- Institute of Digital Health, University Hospital Augsburg, Faculty of Medicine, University of Augsburg, 86356 Neusaess, Germany
- Computer-Aided Medical Procedures and Augmented Reality, School of Computation, Information and Technology, Technical University of Munich, 85748 Garching bei Muenchen, Germany
| | - Florian Schwarz
- Centre for Diagnostic Imaging and Interventional Therapy, Donau-Isar-Klinikum, 94469 Deggendorf, Germany;
| | - Thomas Kroencke
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, 86156 Augsburg, Germany; (S.B.); (L.C.); (L.-M.F.); (A.H.); (J.A.D.); (K.T.); (J.B.); (C.W.); (C.S.-M.); (T.W.)
- Centre for Advanced Analytics and Predictive Sciences (CAAPS), University of Augsburg, 86159 Augsburg, Germany
| |
Collapse
|
8
|
Awais M, Khan N, Khan AK, Rehman A. CT texture analysis for differentiating between peritoneal carcinomatosis and peritoneal tuberculosis: a cross-sectional study. Abdom Radiol (NY) 2024; 49:857-867. [PMID: 37996544 DOI: 10.1007/s00261-023-04103-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
PURPOSE Peritoneal carcinomatosis (PC) and peritoneal tuberculosis (PTB) have similar clinical and radiologic imaging features, which make it very difficult to differentiate between the two entities clinically. Our aim was to determine if the CT textural parameters of omental lesions among patients with PC were different from those with PTB. METHODS All patients who had undergone omental biopsy at our institution from January 2010 to December 2018 and had a tissue diagnosis of PC or PTB were eligible for inclusion. Patients who did not have a contrast-enhanced CT abdomen within one month of the omental biopsy were excluded. A region of interest (ROI) was manually drawn over omental lesions and radiomic features were extracted using open-source LIFEx software. Statistical analysis was performed to compare mean differences in CT texture parameters between the PC and PTB groups. RESULTS A total of 66 patients were included in the study of which 38 and 28 had PC and PTB, respectively. Omental lesions in patients with PC had higher mean radiodensity (mean difference: +32.4; p = 0.001), higher mean entropy (mean difference: +0.11; p < 0.001), and lower mean energy (mean difference: -0.024; p = 0.001) compared to those in PTB. Additionally, omental lesions in the PC group had lower gray-level co-occurrence matrix (GLCM) homogeneity (mean difference: -0.073; p < 0.001) and higher GLCM dissimilarity (mean difference: +0.480; p < 0.001) as compared to the PTB group. CONCLUSION CT texture parameters of omental lesions differed significantly between patients with PTB and those with PC, which may help clinicians in differentiating between the two entities.
Collapse
Affiliation(s)
- Muhammad Awais
- Department of Radiology, Aga Khan University Hospital, Stadium Road, P.O. Box 3500, Karachi, 74800, Sindh, Pakistan.
| | - Noman Khan
- Department of Radiology, Aga Khan University Hospital, Stadium Road, P.O. Box 3500, Karachi, 74800, Sindh, Pakistan
| | - Ayimen Khalid Khan
- Department of Radiology, Aga Khan University Hospital, Stadium Road, P.O. Box 3500, Karachi, 74800, Sindh, Pakistan
| | - Abdul Rehman
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| |
Collapse
|
9
|
Li J, Ma Y, Yang C, Qiu G, Chen J, Tan X, Zhao Y. Radiomics analysis of R2* maps to predict early recurrence of single hepatocellular carcinoma after hepatectomy. Front Oncol 2024; 14:1277698. [PMID: 38463221 PMCID: PMC10920317 DOI: 10.3389/fonc.2024.1277698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 02/09/2024] [Indexed: 03/12/2024] Open
Abstract
OBJECTIVES This study aimed to evaluate the effectiveness of radiomics analysis with R2* maps in predicting early recurrence (ER) in single hepatocellular carcinoma (HCC) following partial hepatectomy. METHODS We conducted a retrospective analysis involving 202 patients with surgically confirmed single HCC having undergone preoperative magnetic resonance imaging between 2018 and 2021 at two different institutions. 126 patients from Institution 1 were assigned to the training set, and 76 patients from Institution 2 were assigned to the validation set. A least absolute shrinkage and selection operator (LASSO) regularization was conducted to operate a logistic regression, then features were identified to construct a radiomic score (Rad-score). Uni- and multi-variable tests were used to assess the correlations of clinicopathological features and Rad-score with ER. We then established a combined model encompassing the optimal Rad-score and clinical-pathological risk factors. Additionally, we formulated and validated a predictive nomogram for predicting ER in HCC. The nomogram's discrimination, calibration, and clinical utility were thoroughly evaluated. RESULTS Multivariable logistic regression revealed the Rad-score, microvascular invasion (MVI), and α fetoprotein (AFP) level > 400 ng/mL as significant independent predictors of ER in HCC. We constructed a nomogram based on these significant factors. The areas under the receiver operator characteristic curve of the nomogram and precision-recall curve were 0.901 and 0.753, respectively, with an F1 score of 0.831 in the training set. These values in the validation set were 0.827, 0.659, and 0.808. CONCLUSION The nomogram that integrates the radiomic score, MVI, and AFP demonstrates high predictive efficacy for estimating the risk of ER in HCC. It facilitates personalized risk classification and therapeutic decision-making for HCC patients.
Collapse
Affiliation(s)
- Jia Li
- Department of Oncology, Central People’s Hospital of Zhanjiang, Zhanjiang, China
| | - Yunhui Ma
- Department of Oncology, Central People’s Hospital of Zhanjiang, Zhanjiang, China
| | - Chunyu Yang
- Department of Radiology, The First School of Clinical Medicine, Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Ganbin Qiu
- Imaging Department of Zhaoqing Medical College, Zhaoqing, China
| | - Jingmu Chen
- Department of Radiology, Central People’s Hospital of Zhanjiang, Zhanjiang, China
| | - Xiaoliang Tan
- Department of Radiology, Central People’s Hospital of Zhanjiang, Zhanjiang, China
| | - Yue Zhao
- Department of Radiology, Central People’s Hospital of Zhanjiang, Zhanjiang, China
| |
Collapse
|
10
|
Saleh M, Virarkar M, Mahmoud HS, Wong VK, Gonzalez Baerga CI, Parikh M, Elsherif SB, Bhosale PR. Radiomics analysis with three-dimensional and two-dimensional segmentation to predict survival outcomes in pancreatic cancer. World J Radiol 2023; 15:304-314. [PMID: 38058604 PMCID: PMC10696186 DOI: 10.4329/wjr.v15.i11.304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/20/2023] [Accepted: 10/23/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Radiomics can assess prognostic factors in several types of tumors, but considering its prognostic ability in pancreatic cancer has been lacking. AIM To evaluate the performance of two different radiomics software in assessing survival outcomes in pancreatic cancer patients. METHODS We retrospectively reviewed pretreatment contrast-enhanced dual-energy computed tomography images from 48 patients with biopsy-confirmed pancreatic ductal adenocarcinoma who later underwent neoadjuvant chemoradiation and surgery. Tumors were segmented using TexRad software for 2-dimensional (2D) analysis and MIM software for 3D analysis, followed by radiomic feature extraction. Cox proportional hazard modeling correlated texture features with overall survival (OS) and progression-free survival (PFS). Cox regression was used to detect differences in OS related to pretreatment tumor size and residual tumor following treatment. The Wilcoxon test was used to show the relationship between tumor volume and the percent of residual tumor. Kaplan-Meier analysis was used to compare survival in patients with different tumor densities in Hounsfield units for both 2D and 3D analysis. RESULTS 3D analysis showed that higher mean tumor density [hazard ratio (HR) = 0.971, P = 0.041)] and higher median tumor density (HR = 0.970, P = 0.037) correlated with better OS. 2D analysis showed that higher mean tumor density (HR = 0.963, P = 0.014) and higher mean positive pixels (HR = 0.962, P = 0.014) correlated with better OS; higher skewness (HR = 3.067, P = 0.008) and higher kurtosis (HR = 1.176, P = 0.029) correlated with worse OS. Higher entropy correlated with better PFS (HR = 0.056, P = 0.036). Models determined that patients with increased tumor size greater than 1.35 cm were likely to have a higher percentage of residual tumors of over 10%. CONCLUSION Several radiomics features can be used as prognostic tools for pancreatic cancer. However, results vary between 2D and 3D analyses. Mean tumor density was the only variable that could reliably predict OS, irrespective of the analysis used.
Collapse
Affiliation(s)
- Mohammed Saleh
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Mayur Virarkar
- Department of Diagnostic Radiology, The University of Florida College of Medicine, Jacksonville, FL 32209, United States
| | - Hagar S Mahmoud
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Vincenzo K Wong
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Carlos Ignacio Gonzalez Baerga
- Department of Diagnostic Radiology, The University of Florida College of Medicine, Jacksonville, FL 32209, United States
| | - Miti Parikh
- Keck School of Medicine, University of South California, Los Angeles, CA 90033, United States
| | - Sherif B Elsherif
- Department of Diagnostic Radiology, The University of Florida College of Medicine, Jacksonville, FL 32209, United States
| | - Priya R Bhosale
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
11
|
Litjens G, Broekmans JPEA, Boers T, Caballo M, van den Hurk MHF, Ozdemir D, van Schaik CJ, Janse MHA, van Geenen EJM, van Laarhoven CJHM, Prokop M, de With PHN, van der Sommen F, Hermans JJ. Computed Tomography-Based Radiomics Using Tumor and Vessel Features to Assess Resectability in Cancer of the Pancreatic Head. Diagnostics (Basel) 2023; 13:3198. [PMID: 37892019 PMCID: PMC10606005 DOI: 10.3390/diagnostics13203198] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/01/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
The preoperative prediction of resectability pancreatic ductal adenocarcinoma (PDAC) is challenging. This retrospective single-center study examined tumor and vessel radiomics to predict the resectability of PDAC in chemo-naïve patients. The tumor and adjacent arteries and veins were segmented in the portal-venous phase of contrast-enhanced CT scans, and radiomic features were extracted. Features were selected via stability and collinearity testing, and least absolute shrinkage and selection operator application (LASSO). Three models, using tumor features, vessel features, and a combination of both, were trained with the training set (N = 86) to predict resectability. The results were validated with the test set (N = 15) and compared to the multidisciplinary team's (MDT) performance. The vessel-features-only model performed best, with an AUC of 0.92 and sensitivity and specificity of 97% and 73%, respectively. Test set validation showed a sensitivity and specificity of 100% and 88%, respectively. The combined model was as good as the vessel model (AUC = 0.91), whereas the tumor model showed poor performance (AUC = 0.76). The MDT's prediction reached a sensitivity and specificity of 97% and 84% for the training set and 88% and 100% for the test set, respectively. Our clinician-independent vessel-based radiomics model can aid in predicting resectability and shows performance comparable to that of the MDT. With these encouraging results, improved, automated, and generalizable models can be developed that reduce workload and can be applied in non-expert hospitals.
Collapse
Affiliation(s)
- Geke Litjens
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Joris P. E. A. Broekmans
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Tim Boers
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Marco Caballo
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Maud H. F. van den Hurk
- Department of Plastic and Reconstructive Surgery, Saint Vincent’s University Hospital, D04 T6F4 Dublin, Ireland
| | - Dilek Ozdemir
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Caroline J. van Schaik
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Markus H. A. Janse
- Image Sciences Institute, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Erwin J. M. van Geenen
- Department of Gastroenterology and Hepatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Cees J. H. M. van Laarhoven
- Department of Surgery, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Mathias Prokop
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Peter H. N. de With
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Fons van der Sommen
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - John J. Hermans
- Department of Medical Imaging, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
12
|
Sijithra PC, Santhi N, Ramasamy N. A review study on early detection of pancreatic ductal adenocarcinoma using artificial intelligence assisted diagnostic methods. Eur J Radiol 2023; 166:110972. [PMID: 37454557 DOI: 10.1016/j.ejrad.2023.110972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, chemo-refractory and recalcitrant cancer and increases the number of deaths. With just around 1 in 4 individuals having respectable tumours, PDAC is frequently discovered when it is in an advanced stage. Accordingly, ED of PDAC improves patient survival. Subsequently, this paper reviews the early detection of PDAC, initially, the work presented an overview of PDAC. Subsequently, it reviews the molecular biology of pancreatic cancer and the development of molecular biomarkers are represented. This article illustrates the importance of identifying PDCA, the Immune Microenvironment of Pancreatic Cancer. Consequently, in this review, traditional and non-traditional imaging techniques are elucidated, traditional and non-traditional methods like endoscopic ultrasound, Multidetector CT, CT texture analysis, PET-CT, magnetic resonance imaging, diffusion-weighted imaging, secondary signs of pancreatic cancer, and molecular imaging. The use of artificial intelligence in pancreatic cancer, novel MRI techniques, and the future directions of AI for PDAC detection and prognosis is then described. Additionally, the research problem definition and motivation, current trends and developments, state of art of survey, and objective of the research are demonstrated in the review. Consequently, this review concluded that Artificial Intelligence Assisted Diagnostic Methods with MRI images can be proposed in future to improve the specificity and the sensitivity of the work, and to classify malignant PDAC with greater accuracy.
Collapse
Affiliation(s)
- P C Sijithra
- Department of Electronics and Communication Engineering, Noorul Islam Centre for Higher Education, Kanyakumari District, Tamilnadu, India.
| | - N Santhi
- Department of Electronics and Communication Engineering, Noorul Islam Centre for Higher Education, Kanyakumari District, Tamilnadu, India
| | - N Ramasamy
- Department of Mechanical Engineering, Noorul Islam Centre for Higher Education, Kanyakumari District, Tamilnadu, India
| |
Collapse
|
13
|
Ahmed TM, Kawamoto S, Hruban RH, Fishman EK, Soyer P, Chu LC. A primer on artificial intelligence in pancreatic imaging. Diagn Interv Imaging 2023; 104:435-447. [PMID: 36967355 DOI: 10.1016/j.diii.2023.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 06/18/2023]
Abstract
Artificial Intelligence (AI) is set to transform medical imaging by leveraging the vast data contained in medical images. Deep learning and radiomics are the two main AI methods currently being applied within radiology. Deep learning uses a layered set of self-correcting algorithms to develop a mathematical model that best fits the data. Radiomics converts imaging data into mineable features such as signal intensity, shape, texture, and higher-order features. Both methods have the potential to improve disease detection, characterization, and prognostication. This article reviews the current status of artificial intelligence in pancreatic imaging and critically appraises the quality of existing evidence using the radiomics quality score.
Collapse
Affiliation(s)
- Taha M Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ralph H Hruban
- Sol Goldman Pancreatic Research Center, Department of Pathology, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Philippe Soyer
- Université Paris Cité, Faculté de Médecine, Department of Radiology, Hôpital Cochin-APHP, 75014, 75006, Paris, France, 7501475006
| | - Linda C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
14
|
Xiang F, He X, Liu X, Li X, Zhang X, Fan Y, Yan S. Development and Validation of a Nomogram for Preoperative Prediction of Early Recurrence after Upfront Surgery in Pancreatic Ductal Adenocarcinoma by Integrating Deep Learning and Radiological Variables. Cancers (Basel) 2023; 15:3543. [PMID: 37509206 PMCID: PMC10377149 DOI: 10.3390/cancers15143543] [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: 05/16/2023] [Revised: 06/29/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Around 80% of pancreatic ductal adenocarcinoma (PDAC) patients experience recurrence after curative resection. We aimed to develop a deep-learning model based on preoperative CT images to predict early recurrence (recurrence within 12 months) in PDAC patients. The retrospective study included 435 patients with PDAC from two independent centers. A modified 3D-ResNet18 network was used for a deep learning model construction. A nomogram was constructed by incorporating deep learning model outputs and independent preoperative radiological predictors. The deep learning model provided the area under the receiver operating curve (AUC) values of 0.836, 0.736, and 0.720 in the development, internal, and external validation datasets for early recurrence prediction, respectively. Multivariate logistic analysis revealed that higher deep learning model outputs (odds ratio [OR]: 1.675; 95% CI: 1.467, 1.950; p < 0.001), cN1/2 stage (OR: 1.964; 95% CI: 1.036, 3.774; p = 0.040), and arterial involvement (OR: 2.207; 95% CI: 1.043, 4.873; p = 0.043) were independent risk factors associated with early recurrence and were used to build an integrated nomogram. The nomogram yielded AUC values of 0.855, 0.752, and 0.741 in the development, internal, and external validation datasets. In conclusion, the proposed nomogram may help predict early recurrence in PDAC patients.
Collapse
Affiliation(s)
- Fei Xiang
- Department of Hepatobiliary Pancreatic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiang He
- Department of Hepatobiliary Surgery I, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xingyu Liu
- Department of Hepatobiliary Pancreatic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xinming Li
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xuchang Zhang
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yingfang Fan
- Department of Hepatobiliary Surgery I, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Sheng Yan
- Department of Hepatobiliary Pancreatic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
15
|
Huang Y, Zhou S, Luo Y, Zou J, Li Y, Chen S, Gao M, Huang K, Lian G. Development and validation of a radiomics model of magnetic resonance for predicting liver metastasis in resectable pancreatic ductal adenocarcinoma patients. Radiat Oncol 2023; 18:79. [PMID: 37165440 PMCID: PMC10170860 DOI: 10.1186/s13014-023-02273-w] [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: 10/14/2022] [Accepted: 04/27/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Nearly one fourth of patients with pancreatic ductal adenocarcinoma (PDAC) occur to liver metastasis after surgery, and liver metastasis is a risk factor for prognosis for those patients with surgery therapy. However, there is no effective way to predict liver metastasis post-operation. METHOD Clinical data and preoperative magnetic resonance imaging (MRI) of PDAC patients diagnosed between July 2010 and July 2020 were retrospectively collected from three hospital centers in China. The significant MRI radiomics features or clinicopathological characteristics were used to establish a model to predict liver metastasis in the development and validation cohort. RESULTS A total of 204 PDAC patients from three hospital centers were divided randomly (7:3) into development and validation cohort. Due to poor predictive value of clinical features, MRI radiomics model had similar receiver operating characteristics curve (ROC) value to clinical-radiomics combing model in development cohort (0.878 vs. 0.880, p = 0.897) but better ROC in validation dataset (0.815 vs. 0.732, p = 0.022). Radiomics model got a sensitivity of 0.872/0.750 and a specificity of 0.760/0.822 to predict liver metastasis in development and validation cohort, respectively. Among 54 patients randomly selected with post-operation specimens, fibrosis markers (α-smooth muscle actin) staining was shown to promote radiomics model with ROC value from 0.772 to 0.923 (p = 0.049) to predict liver metastasis. CONCLUSION This study developed and validated an MRI-based radiomics model and showed a good performance in predicting liver metastasis in resectable PDAC patients.
Collapse
Affiliation(s)
- Yuzhou Huang
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Shurui Zhou
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yanji Luo
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Jinmao Zou
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yaqing Li
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Shaojie Chen
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Ming Gao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Kaihong Huang
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Guoda Lian
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
16
|
Truong LUF, Kleiber JC, Durot C, Brenet E, Barbe C, Hoeffel C, Bazin A, Labrousse M, Dubernard X. The study of predictive factors for the evolution of vestibular schwannomas. Eur Arch Otorhinolaryngol 2023; 280:1661-1670. [PMID: 36114332 DOI: 10.1007/s00405-022-07651-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE The primary objective was to determine whether the analysis of textural heterogeneity of vestibular schwannomas on MRI at diagnosis was predictive of their radiological evolutivity. The secondary objective was to determine whether some clinical or radiological factors could also be predictive of growth. METHODS We conducted a pilot, observational and retrospective study of patients with a vestibular schwannoma, initially monitored, between April 2001 and November 2019 within the Oto-Neurosurgical Institute of Champagne Ardenne, Texture analysis was performed on gadolinium injected T1 and CISS T2 MRI sequences and six parameters were extracted: mean greyscale intensity, standard deviation of the greyscale histogram distribution, entropy, mean positive pixels, skewness and kurtosis, which were analysed by the Lasso method, using statistically penalised Cox models. Extrameatal location, tumour necrosis, perceived hearing loss < 2 years with objectified tone audiometry asymmetry, tinnitus at diagnosis, were investigated by the Log-Rank test to obtain univariate survival analyses. RESULTS 78 patients were included and divided into 2 groups: group A comprising 39 "stable patients", and B comprising the remaining 39 "progressive patients". Independent analysis of the texture factors did not predict the growth potential of vestibular schwannomas. Among the clinical or radiological signs of interest, hearing loss < 2 years was identified as a prognostic factor for tumour progression with a significant trend (p = 0.05). CONCLUSIONS This study did not identify an association between texture analysis and vestibular schwannomas growth. Decreased hearing in the 2 years prior to diagnosis appears to predict potential radiological progression.
Collapse
Affiliation(s)
- Le-Uyen France Truong
- Department of Oto-Rhino-Laryngology and Head and neck surgery of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
| | - Jean Charles Kleiber
- Department of Neurosurgery of the CHU of Reims, Hôpital Maison Blanche, 45 rue Cognacq-Jay, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Carole Durot
- Department of Radiology of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Esteban Brenet
- Department of Oto-Rhino-Laryngology and Head and neck surgery of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Coralie Barbe
- Research and Public Health Unit of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Christine Hoeffel
- Department of Radiology of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Arnaud Bazin
- Department of Neurosurgery of the CHU of Reims, Hôpital Maison Blanche, 45 rue Cognacq-Jay, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Marc Labrousse
- Department of Oto-Rhino-Laryngology and Head and neck surgery of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France
| | - Xavier Dubernard
- Department of Oto-Rhino-Laryngology and Head and neck surgery of the CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France.
- Faculty of Medicine, Reims Champagne Ardenne University, 51100, Reims, France.
- Service d'ORL et Chirurgie cervico-faciale, CHU of Reims, Hôpital Robert Debré, Rue du Général Koenig, 51100, Reims, France.
| |
Collapse
|
17
|
Bian Y, Zhou J, Zhu M, Yu J, Zhao H, Fang X, Liu F, Wang T, Li J, Wang L, Lu J, Shao C. Replacing secretin-enhanced MRCP with MRI radiomics model based on a fully automated pancreas segmentation for assessing pancreatic exocrine function in chronic pancreatitis. Eur Radiol 2023; 33:3580-3591. [PMID: 36884086 DOI: 10.1007/s00330-023-09448-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/06/2023] [Accepted: 01/18/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVES To develop and validate a radiomics nomogram based on a fully automated pancreas segmentation to assess pancreatic exocrine function. Furthermore, we aimed to compare the performance of the radiomics nomogram with the pancreatic flow output rate (PFR) and conclude on the replacement of secretin-enhanced magnetic resonance cholangiopancreatography (S-MRCP) by the radiomics nomogram for pancreatic exocrine function assessment. METHODS All participants underwent S-MRCP between April 2011 and December 2014 in this retrospective study. PFR was quantified using S-MRCP. Participants were divided into normal and pancreatic exocrine insufficiency (PEI) groups using the cut-off of 200 µg/L of fecal elastase-1. Two prediction models were developed including the clinical and non-enhanced T1-weighted imaging radiomics model. A multivariate logistic regression analysis was conducted to develop the prediction models. The models' performances were determined based on their discrimination, calibration, and clinical utility. RESULTS A total of 159 participants (mean age [Formula: see text] standard deviation, 45 years [Formula: see text] 14;119 men) included 85 normal and 74 PEI. All the participants were divided into a training set comprising 119 consecutive patients and an independent validation set comprising 40 consecutive patients. The radiomics score was an independent risk factor for PEI (odds ratio = 11.69; p < 0.001). In the validation set, the radiomics nomogram exhibited the highest performance (AUC, 0.92) in PEI prediction, whereas the clinical nomogram and PFR had AUCs of 0.79 and 0.78, respectively. CONCLUSION The radiomics nomogram accurately predicted pancreatic exocrine function and outperformed pancreatic flow output rate on S-MRCP in patients with chronic pancreatitis. KEY POINTS • The clinical nomogram exhibited moderate performance in diagnosing pancreatic exocrine insufficiency. • The radiomics score was an independent risk factor for pancreatic exocrine insufficiency, and every point rise in the rad-score was associated with an 11.69-fold increase in pancreatic exocrine insufficiency risk. • The radiomics nomogram accurately predicted pancreatic exocrine function and outperformed the clinical model and pancreatic flow output rate quantified by secretin-enhanced magnetic resonance cholangiopancreatography on MRI in patients with chronic pancreatitis.
Collapse
Affiliation(s)
- Yun Bian
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Jian Zhou
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Mengmeng Zhu
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Jieyu Yu
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Haiyan Zhao
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Xu Fang
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Fang Liu
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Tiegong Wang
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Jing Li
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Li Wang
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Jianping Lu
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital, Navy Medical University, Changhai Road 168, Shanghai, 200434, China.
| |
Collapse
|
18
|
Can Imaging Using Radiomics and Fat Fraction Analysis Detect Early Tissue Changes on Historical CT Scans in the Regions of the Pancreas Gland That Subsequently Develop Adenocarcinoma? Diagnostics (Basel) 2023; 13:diagnostics13050941. [PMID: 36900085 PMCID: PMC10001321 DOI: 10.3390/diagnostics13050941] [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: 01/31/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Despite a growing number of effective therapeutic options for patients with pancreatic adenocarcinoma, the prognosis remains dismal mostly due to the late-stage presentation and spread of the cancer to other organs. Because a genomic analysis of pancreas tissue revealed that it may take years, if not decades, for pancreatic cancer to develop, we performed radiomics and fat fraction analysis on contrast-enhanced CT (CECT) scans of patients with historical scans showing no evidence of cancer but who subsequently went on to develop pancreas cancer years later, in an attempt to identify specific imaging features of the normal pancreas that may portend the subsequent development of the cancer. In this IRB-exempt, retrospective, single institution study, CECT chest, abdomen, and pelvis (CAP) scans of 22 patients who had evaluable historical imaging data were analyzed. The images from the "healthy" pancreas were obtained between 3.8 and 13.9 years before the diagnosis of pancreas cancer was established. Afterwards, the images were used to divide and draw seven regions of interest (ROIs) around the pancreas (uncinate, head, neck-genu, body (proximal, middle, and distal) and tail). Radiomic analysis on these pancreatic ROIs consisted of first order quantitative texture analysis features such as kurtosis, skewness, and fat quantification. Of all the variables tested, fat fraction in the pancreas tail (p = 0.029) and asymmetry of the histogram frequency curve (skewness) of pancreas tissue (p = 0.038) were identified as the most important imaging signatures for subsequent cancer development. Changes in the texture of the pancreas as measured on the CECT of patients who developed pancreas cancer years later could be identified, confirming the utility of radiomics-based imaging as a potential predictor of oncologic outcomes. Such findings may be potentially useful in the future to screen patients for pancreatic cancer, thereby helping detect pancreas cancer at an early stage and improving survival.
Collapse
|
19
|
Jamshidi N, Senthilvelan J, Dawson DW, Donahue TR, Kuo MD. Construction of a radiogenomic association map of pancreatic ductal adenocarcinoma. BMC Cancer 2023; 23:189. [PMID: 36843111 PMCID: PMC9969670 DOI: 10.1186/s12885-023-10658-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 02/17/2023] [Indexed: 02/28/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PDAC) persists as a malignancy with high morbidity and mortality that can benefit from new means to characterize and detect these tumors, such as radiogenomics. In order to address this gap in the literature, constructed a transcriptomic-CT radiogenomic (RG) map for PDAC. METHODS In this Institutional Review Board approved study, a cohort of subjects (n = 50) with gene expression profile data paired with histopathologically confirmed resectable or borderline resectable PDAC were identified. Studies with pre-operative contrast-enhanced CT images were independently assessed for a set of 88 predefined imaging features. Microarray gene expression profiling was then carried out on the histopathologically confirmed pancreatic adenocarcinomas and gene networks were constructed using Weighted Gene Correlation Network Analysis (WCGNA) (n = 37). Data were analyzed with bioinformatics analyses, multivariate regression-based methods, and Kaplan-Meier survival analyses. RESULTS Survival analyses identified multiple features of interest that were significantly associated with overall survival, including Tumor Height (P = 0.014), Tumor Contour (P = 0.033), Tumor-stroma Interface (P = 0.014), and the Tumor Enhancement Ratio (P = 0.047). Gene networks for these imaging features were then constructed using WCGNA and further annotated according to the Gene Ontology (GO) annotation framework for a biologically coherent interpretation of the imaging trait-associated gene networks, ultimately resulting in a PDAC RG CT-transcriptome map composed of 3 stage-independent imaging traits enriched in metabolic processes, telomerase activity, and podosome assembly (P < 0.05). CONCLUSIONS A CT-transcriptomic RG map for PDAC composed of semantic and quantitative traits with associated biology processes predictive of overall survival, was constructed, that serves as a reference for further mechanistic studies for non-invasive phenotyping of pancreatic tumors.
Collapse
Affiliation(s)
- Neema Jamshidi
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, 757 Westwood Ave, Suite 2125, Los Angeles, CA, 90095, USA. .,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
| | - Jayasuriya Senthilvelan
- grid.19006.3e0000 0000 9632 6718Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, 757 Westwood Ave, Suite 2125, Los Angeles, CA 90095 USA
| | - David W. Dawson
- grid.19006.3e0000 0000 9632 6718Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Pathology, University of California, Los Angeles, CA USA
| | - Timothy R. Donahue
- grid.19006.3e0000 0000 9632 6718Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Surgical Oncology, University of California, Los Angeles, CA USA
| | - Michael D. Kuo
- grid.194645.b0000000121742757Medical AI Laboratory Program, The University of Hong Kong, Hong Kong SAR, Hong Kong
| |
Collapse
|
20
|
Radiomics analysis of contrast-enhanced T1W MRI: predicting the recurrence of acute pancreatitis. Sci Rep 2023; 13:2762. [PMID: 36797285 PMCID: PMC9935887 DOI: 10.1038/s41598-022-13650-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 05/26/2022] [Indexed: 02/18/2023] Open
Abstract
To investigate the predictive value of radiomics based on T1-weighted contrast-enhanced MRI (CE-MRI) in forecasting the recurrence of acute pancreatitis (AP). A total of 201 patients with first-episode of acute pancreatitis were enrolled retrospectively (140 in the training cohort and 61 in the testing cohort), with 69 and 30 patients who experienced recurrence in each cohort, respectively. Quantitative image feature extraction was obtained from MR contrast-enhanced late arterial-phase images. The optimal radiomics features retained after dimensionality reduction were used to construct the radiomics model through logistic regression analysis, and the clinical characteristics were collected to construct the clinical model. The nomogram model was established by linearly integrating the clinically independent risk factor with the optimal radiomics signature. The five best radiomics features were determined by dimensionality reduction. The radiomics model had a higher area under the receiver operating characteristic curve (AUC) than the clinical model for estimating the recurrence of acute pancreatitis for both the training cohort (0.915 vs. 0.811, p = 0.020) and testing cohort (0.917 vs. 0.681, p = 0.002). The nomogram model showed good performance, with an AUC of 0.943 in the training cohort and 0.906 in the testing cohort. The radiomics model based on CE-MRI showed good performance for optimizing the individualized prediction of recurrent acute pancreatitis, which provides a reference for the prevention and treatment of recurrent pancreatitis.
Collapse
|
21
|
Tikhonova VS, Karmazanovsky GG, Kondratyev EV, Gruzdev IS, Mikhaylyuk KA, Sinelnikov MY, Revishvili AS. Radiomics model-based algorithm for preoperative prediction of pancreatic ductal adenocarcinoma grade. Eur Radiol 2023; 33:1152-1161. [PMID: 35986774 DOI: 10.1007/s00330-022-09046-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/24/2022] [Accepted: 07/13/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To develop diagnostic radiomic model-based algorithm for pancreatic ductal adenocarcinoma (PDAC) grade prediction. METHODS Ninety-one patients with histologically confirmed PDAC and preoperative CT were divided into subgroups based on tumor grade. Two histology-blinded radiologists independently segmented lesions for quantitative texture analysis in all contrast enhancement phases. The ratio of densities of PDAC and unchanged pancreatic tissue, and relative tumor enhancement (RTE) in arterial, portal venous, and delayed phases of the examination were calculated. Principal component analysis was used for multivariate predictor analysis. The selection of predictors in the binary logistic model was carried out in 2 stages: (1) using one-factor logistic models (selection criterion was p < 0.1); (2) using regularization (LASSO regression after standardization of variables). Predictors were included in proportional odds models without interactions. RESULTS There were significant differences in 4, 16, and 8 texture features out of 62 for the arterial, portal venous, and delayed phases of the study, respectively (p < 0.1). After selection, the final diagnostic model included such radiomics features as DISCRETIZED HU standard, DISCRETIZED HUQ3, GLCM Correlation, GLZLM LZLGE for the portal venous phase of the contrast enhancement, and CONVENTIONAL_HUQ3 for the delayed phase of CT study. On its basis, a diagnostic model was built, showing AUC for grade ≥ 2 of 0.75 and AUC for grade 3 of 0.66. CONCLUSION Radiomics features vary in PDAC of different grades and increase the accuracy of CT in preoperative diagnosis. We have developed a diagnostic model, including texture features, which can be used to predict the grade of PDAC. KEY POINTS • A diagnostic algorithm based on CT texture features for preoperative PDAC grade prediction was developed. • The assumption that the scanning protocol can influence the results of texture analysis was confirmed and assessed. • Our results show that tumor differentiation grade can be assessed with sufficient diagnostic accuracy using CT texture analysis presented in this study.
Collapse
Affiliation(s)
| | - Grigory G Karmazanovsky
- A.V. Vishnevsky National Medical Research Centre of Surgery, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Ivan S Gruzdev
- A.V. Vishnevsky National Medical Research Centre of Surgery, Moscow, Russia
| | | | - Mikhail Y Sinelnikov
- Research Institute of Human Morphology, Moscow, Russia.
- Sechenov University, Moscow, Russia.
| | | |
Collapse
|
22
|
Lu J, Jiang N, Zhang Y, Li D. A CT based radiomics nomogram for differentiation between focal-type autoimmune pancreatitis and pancreatic ductal adenocarcinoma. Front Oncol 2023; 13:979437. [PMID: 36937433 PMCID: PMC10014827 DOI: 10.3389/fonc.2023.979437] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 02/20/2023] [Indexed: 03/05/2023] Open
Abstract
Objectives The purpose of this study was to develop and validate an CT-based radiomics nomogram for the preoperative differentiation of focal-type autoimmune pancreatitis from pancreatic ductal adenocarcinoma. Methods 96 patients with focal-type autoimmune pancreatitis and pancreatic ductal adenocarcinoma have been enrolled in the study (32 and 64 cases respectively). All cases have been confirmed by imaging, clinical follow-up and/or pathology. The imaging data were considered as: 70% training cohort and 30% test cohort. Pancreatic lesions have been manually delineated by two radiologists and image segmentation was performed to extract radiomic features from the CT images. Independent-sample T tests and LASSO regression were used for feature selection. The training cohort was classified using a variety of machine learning-based classifiers, and 5-fold cross-validation has been performed. The classification performance was evaluated using the test cohort. Multivariate logistic regression analysis was then used to develop a radiomics nomogram model, containing the CT findings and Rad-Score. Calibration curves have been plotted showing the agreement between the predicted and actual probabilities of the radiomics nomogram model. Different patients have been selected to test and evaluate the model prediction process. Finally, receiver operating characteristic curves and decision curves were plotted, and the radiomics nomogram model was compared with a single model to visually assess its diagnostic ability. Results A total of 158 radiomics features were extracted from each image. 7 features were selected to construct the radiomics model, then a variety of classifiers were used for classification and multinomial logistic regression (MLR) was selected to be the optimal classifier. Combining CT findings with radiomics model, a prediction model based on CT findings and radiomics was finally obtained. The nomogram model showed a good sensitivity and specificity with AUCs of 0.87 and 0.83 in training and test cohorts, respectively. The areas under the curve and decision curve analysis showed that the radiomics nomogram model may provide better diagnostic performance than the single model and achieve greater clinical net benefits than the CT finding model and radiomics signature model individually. Conclusions The CT image-based radiomics nomogram model can accurately distinguish between focal-type autoimmune pancreatitis and pancreatic ductal adenocarcinoma patients and provide additional clinical benefits.
Collapse
Affiliation(s)
- Jia Lu
- Department of Radiology, The People’s Hospital of China Medical University and The People’s Hospital of Liaoning Province, Shenyang, China
| | - Nannan Jiang
- Department of Radiology, The People’s Hospital of Liaoning Province, Shenyang, China
| | - Yuqing Zhang
- Department of Radiology, The People’s Hospital of China Medical University and The People’s Hospital of Liaoning Province, Shenyang, China
| | - Daowei Li
- Department of Radiology, The People’s Hospital of China Medical University and The People’s Hospital of Liaoning Province, Shenyang, China
- *Correspondence: Daowei Li,
| |
Collapse
|
23
|
Wang F, Zhao Y, Xu J, Shao S, Yu D. Development and external validation of a radiomics combined with clinical nomogram for preoperative prediction prognosis of resectable pancreatic ductal adenocarcinoma patients. Front Oncol 2022; 12:1037672. [PMID: 36518321 PMCID: PMC9742428 DOI: 10.3389/fonc.2022.1037672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2023] Open
Abstract
PURPOSE To develop and externally validate a prognosis nomogram based on contrast-enhanced computed tomography (CECT) combined clinical for preoperative prognosis prediction of patients with pancreatic ductal adenocarcinoma (PDAC). METHODS 184 patients from Center A with histopathologically confirmed PDAC who underwent CECT were included and allocated to training cohort (n=111) and internal validation cohort (n=28). The radiomic score (Rad - score) for predicting overall survival (OS) was constructed by using the least absolute shrinkage and selection operator (LASSO). Univariate and multivariable Cox regression analysis was used to construct clinic-pathologic features. Finally, a radiomics nomogram incorporating the Rad - score and clinical features was established. External validation was performed using Center B dataset (n = 45). The validation of nomogram was evaluated by calibration curve, Harrell's concordance index (C-index) and decision curve analysis (DCA). The Kaplan-Meier (K-M) method was used for OS analysis. RESULTS Univariate and multivariate analysis indicated that Rad - score, preoperative CA 19-9 and postoperative American Joint Committee on Cancer (AJCC) TNM stage were significant prognostic factors. The nomogram based on Rad - score and preoperative CA19-9 was found to exhibit excellent prediction ability: in the training cohort, C-index was superior to that of the preoperative CA19-9 (0.713 vs 0.616, P< 0.001) and AJCC TNM stage (0.713 vs 0.614, P< 0.001); the C-index was also had good performance in the validation cohort compared with CA19-9 (internal validation cohort: 0.694 vs 0.555, P< 0.001; external validation cohort: 0.684 vs 0.607, P< 0.001) and AJCC TNM stage (internal validation cohort: 0.694 vs 0.563, P< 0.001; external validation cohort: 0.684 vs 0.596, P< 0.001). The calibration plot and DCA showed excellent predictive accuracy in the validation cohort. CONCLUSION We established a well-designed nomogram to accurately predict OS of PDAC preoperatively. The nomogram showed a satisfactory prediction effect and was worthy of further evaluation in the future.
Collapse
Affiliation(s)
- Fangqing Wang
- Departments of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuxuan Zhao
- Departments of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jianwei Xu
- Department of Pancreatic Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Sai Shao
- Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Dexin Yu
- Departments of Radiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
24
|
Yang J, Liu Y, Liu S. Comment on "Prognostic value of preoperative enhanced computed tomography as a quantitative imaging biomarker in pancreatic cancer". World J Gastroenterol 2022; 28:6310-6313. [PMID: 36504551 PMCID: PMC9730437 DOI: 10.3748/wjg.v28.i44.6310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/26/2022] [Accepted: 11/16/2022] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies because of its high invasiveness and metastatic potential. Computed tomography (CT) is often used as a preliminary diagnostic tool for pancreatic cancer, and it is increasingly used to predict treatment response and disease stage. Recently, a study published in World Journal of Gastroenterology reported that quantitative analysis of preoperative enhanced CT data can be used to predict postoperative overall survival in patients with PDAC. A tumor relative enhancement ratio of ≤ 0.7 indicates a higher tumor stage and poor prognosis.
Collapse
Affiliation(s)
- Jian Yang
- Central Laboratory, The Third Affiliated Hospital, Qiqihar Medical University, Qiqihar 161000, Heilongjiang Province, China
| | - Ying Liu
- Department of Medical Oncology, The Third Affiliated Hospital, Qiqihar Medical University, Qiqihar 161000, Heilongjiang Province, China
| | - Shi Liu
- Central Laboratory, The Third Affiliated Hospital, Qiqihar Medical University, Qiqihar 161000, Heilongjiang Province, China
| |
Collapse
|
25
|
Quantitative Radiomic Features From Computed Tomography Can Predict Pancreatic Cancer up to 36 Months Before Diagnosis. Clin Transl Gastroenterol 2022; 14:e00548. [PMID: 36434803 PMCID: PMC9875961 DOI: 10.14309/ctg.0000000000000548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/18/2022] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Pancreatic cancer is the third leading cause of cancer deaths among men and women in the United States. We aimed to detect early changes on computed tomography (CT) images associated with pancreatic ductal adenocarcinoma (PDAC) based on quantitative imaging features (QIFs) for patients with and without chronic pancreatitis (CP). METHODS Adults 18 years and older diagnosed with PDAC in 2008-2018 were identified. Their CT scans 3 months-3 years before the diagnosis date were matched to up to 2 scans of controls. The pancreas was automatically segmented using a previously developed algorithm. One hundred eleven QIFs were extracted. The data set was randomly split for training/validation. Neighborhood and principal component analyses were applied to select the most important features. A conditional support vector machine was used to develop prediction algorithms separately for patients with and without CP. The computer labels were compared with manually reviewed CT images 2-3 years before the index date in 19 cases and 19 controls. RESULTS Two hundred twenty-seven of 554 scans of non-CP cancer cases/controls and 70 of 140 scans of CP cancer cases/controls were included (average age 71 and 68 years, 51% and 44% females for non-CP patients and patients with CP, respectively). The QIF-based algorithms varied based on CP status. For non-CP patients, accuracy measures were 94%-95% and area under the curve (AUC) measures were 0.98-0.99. Sensitivity, specificity, positive predictive value, and negative predictive value were in the ranges of 88%-91%, 96%-98%, 91%-95%, and 94%-96%, respectively. QIFs on CT examinations within 2-3 years before the index date also had very high predictive accuracy (accuracy 95%-98%; AUC 0.99-1.00). The QIF-based algorithm outperformed manual rereview of images for determination of PDAC risk. For patients with CP, the algorithms predicted PDAC perfectly (accuracy 100% and AUC 1.00). DISCUSSION QIFs can accurately predict PDAC for both non-CP patients and patients with CP on CT imaging and represent promising biomarkers for early detection of pancreatic cancer.
Collapse
|
26
|
Kаrmаzаnovsky GG, Kondratyev EV, Gruzdev IS, Tikhonova VS, Shantarevich MY, Zamyatina KA, Stashkiv VI, Revishvili AS. Modern Radiation Diagnostics and Intelligent Personalized Technologies in Hepatopancreatology. ANNALS OF THE RUSSIAN ACADEMY OF MEDICAL SCIENCES 2022; 77:245-253. [DOI: 10.15690/vramn2053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Timely instrumental diagnosis of diseases of the hepatopancreatoduodenal region, especially of an oncological nature, is the key to successful treatment, improving prognosis and improving the quality of life of patients. At the moment, the possibilities of radiation diagnostics make it possible to identify and evaluate the nature of the blood supply to the neoplasm, its prevalence, cellularity, and in the case of MRI studies with hepatospecific contrast agents, also evaluate the functional activity of liver cells. Nevertheless, the steady development of methods for treating cancer patients, in particular, chemotherapy, and a personalized approach to the choice of patient management tactics require a detailed assessment of the morphological types of certain neoplasms. The need for dynamic monitoring of the results of treatment, monitoring of accidentally detected, potentially malignant neoplasms, and the development of screening programs determine the steady increase in the number of CT and MR examinations performed annually in the world and in our country. These factors have led to the application of texture analysis or radiomics and machine learning algorithms. At the same time, such techniques as radiography, ultrasound, CT and MRI with extracellular and tissue-specific contrast enhancement, and MRI-DWI do not lose their significance. The ongoing research allows the Federal State Budgetary Institution National Medical Research Center of Surgery named after A.V. Vishnevsky of the Ministry of Health of Russia to implement the concept of preoperative non-invasive diagnosis and differential diagnosis of surgical and oncological diseases of the hepatopancreatoduodenal region and apply the knowledge gained in planning surgical treatment. Implementation of the problem of post-processor data processing of radiation diagnostics of surgical and oncological diseases of the hepatopancreatoduodenal region using radiomics and AI technologies is important and extremely relevant for modern medicine.
Collapse
|
27
|
Mukherjee S, Patra A, Khasawneh H, Korfiatis P, Rajamohan N, Suman G, Majumder S, Panda A, Johnson MP, Larson NB, Wright DE, Kline TL, Fletcher JG, Chari ST, Goenka AH. Radiomics-based Machine-learning Models Can Detect Pancreatic Cancer on Prediagnostic Computed Tomography Scans at a Substantial Lead Time Before Clinical Diagnosis. Gastroenterology 2022; 163:1435-1446.e3. [PMID: 35788343 DOI: 10.1053/j.gastro.2022.06.066] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND & AIMS Our purpose was to detect pancreatic ductal adenocarcinoma (PDAC) at the prediagnostic stage (3-36 months before clinical diagnosis) using radiomics-based machine-learning (ML) models, and to compare performance against radiologists in a case-control study. METHODS Volumetric pancreas segmentation was performed on prediagnostic computed tomography scans (CTs) (median interval between CT and PDAC diagnosis: 398 days) of 155 patients and an age-matched cohort of 265 subjects with normal pancreas. A total of 88 first-order and gray-level radiomic features were extracted and 34 features were selected through the least absolute shrinkage and selection operator-based feature selection method. The dataset was randomly divided into training (292 CTs: 110 prediagnostic and 182 controls) and test subsets (128 CTs: 45 prediagnostic and 83 controls). Four ML classifiers, k-nearest neighbor (KNN), support vector machine (SVM), random forest (RM), and extreme gradient boosting (XGBoost), were evaluated. Specificity of model with highest accuracy was further validated on an independent internal dataset (n = 176) and the public National Institutes of Health dataset (n = 80). Two radiologists (R4 and R5) independently evaluated the pancreas on a 5-point diagnostic scale. RESULTS Median (range) time between prediagnostic CTs of the test subset and PDAC diagnosis was 386 (97-1092) days. SVM had the highest sensitivity (mean; 95% confidence interval) (95.5; 85.5-100.0), specificity (90.3; 84.3-91.5), F1-score (89.5; 82.3-91.7), area under the curve (AUC) (0.98; 0.94-0.98), and accuracy (92.2%; 86.7-93.7) for classification of CTs into prediagnostic versus normal. All 3 other ML models, KNN, RF, and XGBoost, had comparable AUCs (0.95, 0.95, and 0.96, respectively). The high specificity of SVM was generalizable to both the independent internal (92.6%) and the National Institutes of Health dataset (96.2%). In contrast, interreader radiologist agreement was only fair (Cohen's kappa 0.3) and their mean AUC (0.66; 0.46-0.86) was lower than each of the 4 ML models (AUCs: 0.95-0.98) (P < .001). Radiologists also recorded false positive indirect findings of PDAC in control subjects (n = 83) (7% R4, 18% R5). CONCLUSIONS Radiomics-based ML models can detect PDAC from normal pancreas when it is beyond human interrogation capability at a substantial lead time before clinical diagnosis. Prospective validation and integration of such models with complementary fluid-based biomarkers has the potential for PDAC detection at a stage when surgical cure is a possibility.
Collapse
Affiliation(s)
| | - Anurima Patra
- Department of Radiology, Tata Medical Centre, Kolkata, India
| | - Hala Khasawneh
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Garima Suman
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Shounak Majumder
- Department of Gastroenterology, Mayo Clinic, Rochester, Minnesota
| | - Ananya Panda
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Matthew P Johnson
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Nicholas B Larson
- Department of Radiology, Mayo Clinic, Rochester, Minnesota; Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Suresh T Chari
- Department of Gastroenterology, Mayo Clinic, Rochester, Minnesota; Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
28
|
CT perfusion as a potential biomarker for pancreatic ductal adenocarcinoma during routine staging and restaging. ABDOMINAL RADIOLOGY (NEW YORK) 2022; 47:3770-3781. [PMID: 35972550 DOI: 10.1007/s00261-022-03638-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE To evaluate the significance of CT perfusion parameters predicting response to neoadjuvant therapy in patients with pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS Seventy patients with PDAC prospectively had CT perfusion acquisition incorporated into baseline multiphase staging CT. Twenty-eight who were naïve to therapy were retained for further investigation. Perfusion was performed 5-42.5 s after contrast, followed by parenchymal and portal venous phases. Blood flow (BF), blood volume (BV), and permeability surface area product (PS) were calculated using deconvolution algorithms. Patients were categorized as responders or non-responders per RECIST 1.1. Perfusion variables with AUC ≥ 0.70 in differentiating responders from non-responders were retained. Logistic regression was used to assess associations between baseline perfusion variables and response. RESULTS 18 of 28 patients showed favorable response to therapy. Baseline heterogeneity variables in tumor max ROI were higher in non-responders than responders [median BF coefficient of variation (CV) 0.91 vs. 0.51 respectively, odds ratio (OR) 6.8 per one standard deviation (1-SD) increase, P = 0.047; median PS CV 1.6 vs. 0.68, OR 3.9 per 1-SD increase, P = 0.047; and median BV CV 0.75 vs. 0.54, OR = 4.0 per 1-SD increase, P = 0.047]. Baseline BV mean in tumor center was lower in non-responders than responders (median BV mean: 0.74 vs. 2.9 ml/100 g respectively, OR 0.28 per 1-SD increase, P = 0.047). CONCLUSION For patients with PDAC receiving neoadjuvant therapy, lower and more heterogeneous perfusion parameters correlated with an unfavorable response to therapy. Such quantitative information can be acquired utilizing a comprehensive protocol interleaving perfusion CT acquisition with standard of care multiphase CT scans using a single contrast injection, which could be used to identify surgical candidates and predict outcome.
Collapse
|
29
|
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
|
30
|
Liver metastases in pancreatic ductal adenocarcinoma: a predictive model based on CT texture analysis. Radiol Med 2022; 127:1079-1084. [PMID: 36057929 DOI: 10.1007/s11547-022-01548-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/18/2022] [Indexed: 10/14/2022]
|
31
|
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: 12] [Impact Index Per Article: 4.0] [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
|
32
|
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
|
33
|
Keyl J, Kasper S, Wiesweg M, Götze J, Schönrock M, Sinn M, Berger A, Nasca E, Kostbade K, Schumacher B, Markus P, Albers D, Treckmann J, Schmid KW, Schildhaus HU, Siveke JT, Schuler M, Kleesiek J. Multimodal survival prediction in advanced pancreatic cancer using machine learning. ESMO Open 2022; 7:100555. [PMID: 35988455 PMCID: PMC9588888 DOI: 10.1016/j.esmoop.2022.100555] [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] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/23/2022] Open
Abstract
Background Existing risk scores appear insufficient to assess the individual survival risk of patients with advanced pancreatic ductal adenocarcinoma (PDAC) and do not take advantage of the variety of parameters that are collected during clinical care. Methods In this retrospective study, we built a random survival forest model from clinical data of 203 patients with advanced PDAC. The parameters were assessed before initiation of systemic treatment and included age, CA19-9, C-reactive protein, metastatic status, neutrophil-to-lymphocyte ratio and total serum protein level. Separate models including imaging and molecular parameters were built for subgroups. Results Over the entire cohort, a model based on clinical parameters achieved a c-index of 0.71. Our approach outperformed the American Joint Committee on Cancer (AJCC) staging system and the modified Glasgow Prognostic Score (mGPS) in the identification of high- and low-risk subgroups. Inclusion of the KRAS p.G12D mutational status could further improve the prediction, whereas radiomics data of the primary tumor only showed little benefit. In an external validation cohort of PDAC patients with liver metastases, our model achieved a c-index of 0.67 (mGPS: 0.59). Conclusions The combination of multimodal data and machine-learning algorithms holds potential for personalized prognostication in advanced PDAC already at diagnosis.
We developed a machine-learning-based prediction model that outperforms the AJCC staging system and mGPS. Applying our model to an external validation cohort demonstrates generalizability. Explainable machine learning enables to understand the decision making of our model and identifies relevant parameters. Combining clinical, imaging and genetic data holds potential for personalized prognostication in advanced PDAC.
Collapse
Affiliation(s)
- J Keyl
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany.
| | - S Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - M Wiesweg
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - J Götze
- Department of Internal Medicine II, Oncology, Hematology, BMT and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - M Schönrock
- Department of Internal Medicine II, Oncology, Hematology, BMT and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - M Sinn
- Department of Internal Medicine II, Oncology, Hematology, BMT and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - A Berger
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany
| | - E Nasca
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany
| | - K Kostbade
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - B Schumacher
- Department of Gastroenterology, Elisabeth Hospital Essen, Essen, Germany
| | - P Markus
- Department of General Surgery and Traumatology, Elisabeth Hospital Essen, Essen, Germany
| | - D Albers
- Department of Gastroenterology, Elisabeth Hospital Essen, Essen, Germany
| | - J Treckmann
- Department of General, Visceral and Transplant Surgery, West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
| | - K W Schmid
- Medical Faculty, University of Duisburg-Essen, Essen, Germany; Institute of Pathology, West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
| | - H-U Schildhaus
- Medical Faculty, University of Duisburg-Essen, Essen, Germany; Institute of Pathology, West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
| | - J T Siveke
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany; Bridge Institute of Experimental Tumor Therapy (BIT), West German Cancer Center, University Hospital Essen (AöR), Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK) Partner site Essen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M Schuler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - J Kleesiek
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
34
|
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
|
35
|
A systematic review of prognosis predictive role of radiomics in pancreatic cancer: heterogeneity markers or statistical tricks? Eur Radiol 2022; 32:8443-8452. [PMID: 35904618 DOI: 10.1007/s00330-022-08922-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 04/07/2022] [Accepted: 05/30/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES We aimed to systematically evaluate the prognostic prediction accuracy of radiomics features extracted from pre-treatment imaging in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS Radiomics literature on overall survival (OS) prediction of PDAC were all included in this systematic review. A further meta-analysis was performed on the effect size of first-order entropy. Methodological quality and risk of bias of the included studies were assessed by the radiomics quality score (RQS) and prediction model risk of bias assessment tool (PROBAST). RESULTS Twenty-three studies were finally identified in this review. Two (8.7%) studies compared prognosis prediction ability between radiomics model and TNM staging model by C-index, and both showed a better performance of the radiomics. Twenty-one (91.3%) studies reported significant predictive values of radiomics features. Nine (39.1%) studies were included in the meta-analysis, and it showed a significant correlation between first-order entropy and OS (HR 1.66, 95%CI 1.18-2.34). RQS assessment revealed validation was only performed in 5 (21.7%) studies on internal datasets and 2 (8.7%) studies on external datasets. PROBAST showed that 22 (95.7%) studies have a high risk of bias in participants because of the retrospective study design. CONCLUSION First-order entropy was significantly associated with OS and might improve the accuracy of PDAC prognosis prediction. Existing studies were poorly validated, and it should be noted in future studies. Modification of PROBAST for radiomics studies is necessary since the strict requirements of prospective study design may not be applicable to the demand for a large sample size in the model construction stage. KEY POINTS • Radiomics based on the primary lesion holds great potential for prognosis prediction. First-order entropy was significantly associated with the overall survival of PDAC and might improve the accuracy of current PDAC prognosis prediction. • We strongly recommend that at least an internal validation should be conducted in any radiomics study. Attention should be paid to the complex relationships between radiomics features. • Due to the close relationship between radiomics and big data, the strict requirement of prospective study design in PROABST may not be appropriate for radiomics studies. A balance between study types and sample sizes for radiomics studies needs to be found in the model construction stage.
Collapse
|
36
|
Computed Tomography-based Radiomics Evaluation of Postoperative Local Recurrence of Pancreatic Ductal Adenocarcinoma. Acad Radiol 2022; 30:680-688. [PMID: 35906151 DOI: 10.1016/j.acra.2022.05.019] [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: 03/18/2022] [Revised: 05/24/2022] [Accepted: 05/29/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To develop and validate an effective model for identifying patients with postoperative local disease recurrence of pancreatic ductal adenocarcinoma (PDAC). METHODS A total of 153 patients who had undergone surgical resection of PDAC with regular postoperative follow-up were consecutively enrolled and randomly divided into training (n = 108) and validation (n = 45) cohorts. The postoperative soft-tissue biopsy results or clinical follow-up results served as the reference diagnostic criteria. Radiomics analysis of the postoperative soft-tissue was performed on a commercially available prototype software using portal vein phase image. Three models were built to characterize postoperative soft tissue: computed tomography (CT)-based radiomics, clinicoradiological, and their combination. The area under the receiver operating characteristic curves (AUC) was used to evaluate the differential diagnostic performance. A nomogram was used to select the final model with best performance. One radiologist's diagnostic choices that were made with and without the nomogram's assistance were evaluated. RESULTS A seven-feature-combined radiomics signature was constructed as a predictor of postoperative local recurrence. The nomogram model combining the radiomics signature with postoperative CA 19-9 elevation showed the best performance (training cohort, AUC = 0.791 [95%CI: 0.707, 0.876]; validation cohort, AUC = 0.742 [95%CI: 0.590, 0.894]). In the validation cohort, the AUC for differential diagnosis was significantly improved for the combined model relative to that for postoperative CA 19-9 elevation (AUC = 0.742 vs. 0.533, p < 0.001). The calibration curve and decision curve analysis demonstrated the clinical usefulness of the proposed nomogram. The diagnostic performance of the radiologist was not significantly improve by using the proposed nomogram (AUC = 0.742 vs. 0.670, p = 0.17). CONCLUSION The combined model using CT radiomic features and CA 19-9 elevation effectively characterized postoperative soft tissue and potentially may improve treatment strategies and facilitate personalized treatment for PDAC after surgical resection.
Collapse
|
37
|
Zhou L, Feng F, Yang Y, Zheng X, Yang Y. Prognostic predictors of non-small cell lung cancer treated with curative resection: the role of preoperative CT texture features, clinical features, and laboratory parameters. Clin Radiol 2022; 77:e765-e770. [PMID: 35843728 DOI: 10.1016/j.crad.2022.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/11/2022] [Accepted: 06/15/2022] [Indexed: 11/03/2022]
Abstract
AIM To explore the value of preoperative contrast-enhanced computed tomography (CT) tumour texture characteristics, and clinical and laboratory parameters on the prognosis of curative resection for non-small-cell lung cancer (NSCLC). MATERIALS AND METHODS This retrospective study included 64 patients (34 men and 30 women) with NSCLC who underwent curative resection and were then followed up for 5 years or until death. Preoperative contrast-enhanced CT images, clinical features, and laboratory parameters were collected for these patients. CT texture features of the primary tumour before surgery were extracted from the contrast-enhanced CT images using ImageJ software. Based on the cut-off values determined by X-tile software, the preoperative CT texture features, clinical features, and laboratory parameters were divided into two groups. Kaplan-Meier survival curves and log-rank tests were used to compare the 5-year overall survival (OS) of patients. Multivariate Cox regression analysis was used to determine the independent factors influencing the prognosis. RESULTS The mean survival was 51.5 months. Tumour volume, entropy, platelet-to-lymphocyte ratio (PLR), prognostic nutritional index (PNI), and albumin-to-globulin ratio (AGR) were shown to be significantly associated with 5-year OS (p<0.05). Multivariate Cox regression analysis revealed that entropy was the independent factor of prognosis (hazard ratio 4.375, 95% confidence interval [CI]: 1.646-11.620, p=0.003). CONCLUSION Entropy is an important and potentially non-invasive imaging biomarker for predicting the prognosis of NSCLC undergoing curative resection.
Collapse
Affiliation(s)
- L Zhou
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226361, PR China.
| | - F Feng
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226361, PR China
| | - Y Yang
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226361, PR China
| | - X Zheng
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226361, PR China
| | - Y Yang
- Department of Radiology, Affiliated Tumour Hospital of Nantong University, Nantong, Jiangsu 226361, PR China
| |
Collapse
|
38
|
Zhang Y, Zhuang Y, Ge Y, Wu PY, Zhao J, Wang H, Song B. MRI whole-lesion texture analysis on ADC maps for the prognostic assessment of ischemic stroke. BMC Med Imaging 2022; 22:115. [PMID: 35778678 PMCID: PMC9250246 DOI: 10.1186/s12880-022-00845-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/23/2022] [Indexed: 11/28/2022] Open
Abstract
Background This study aims is to explore whether it is feasible to use magnetic resonance texture analysis (MRTA) in order to distinguish favorable from unfavorable function outcomes and determine the prognostic factors associated with favorable outcomes of stroke. Methods The retrospective study included 103 consecutive patients who confirmed unilateral anterior circulation subacute ischemic stroke by computed tomography angiography between January 2018 and September 2019. Patients were divided into favorable outcome (modified Rankin scale, mRS ≤ 2) and unfavorable outcome (mRS > 2) groups according to mRS scores at day 90. Two radiologists manually segmented the infarction lesions based on diffusion-weighted imaging and transferred the images to corresponding apparent diffusion coefficient (ADC) maps in order to extract texture features. The prediction models including clinical characteristics and texture features were built using multiple logistic regression. A univariate analysis was conducted to assess the performance of the mean ADC value of the infarction lesion. A Delong’s test was used to compare the predictive performance of models through the receiver operating characteristic curve. Results The mean ADC performance was moderate [AUC = 0.60, 95% confidence interval (CI) 0.49–0.71]. The texture feature model of the ADC map (tADC), contained seven texture features, and presented good prediction performance (AUC = 0.83, 95%CI 0.75–0.91). The energy obtained after wavelet transform, and the kurtosis and skewness obtained after Laplacian of Gaussian transformation were identified as independent prognostic factors for the favorable stroke outcomes. In addition, the combination of the tADC model and clinical characteristics (hypertension, diabetes mellitus, smoking, and atrial fibrillation) exhibited a subtly better performance (AUC = 0.86, 95%CI 0.79–0.93; P > 0.05, Delong’s). Conclusion The models based on MRTA on ADC maps are useful to evaluate the clinical function outcomes in patients with unilateral anterior circulation ischemic stroke. Energy obtained after wavelet transform, kurtosis obtained after Laplacian of Gaussian transform, and skewness obtained after Laplacian of Gaussian transform were identified as independent prognostic factors for favorable stroke outcomes.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Radiology, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199, People's Republic of China
| | - Yuzhong Zhuang
- Department of Radiology, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199, People's Republic of China
| | - Yaqiong Ge
- Department of Medicine, GE Healthcare, Shanghai, People's Republic of China
| | - Pu-Yeh Wu
- Department of Medicine, GE Healthcare, Beijing, People's Republic of China
| | - Jing Zhao
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, People's Republic of China
| | - Hao Wang
- Department of Radiology, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199, People's Republic of China.
| | - Bin Song
- Department of Radiology, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199, People's Republic of China.
| |
Collapse
|
39
|
Gao JF, Pan Y, Lin XC, Lu FC, Qiu DS, Liu JJ, Huang HG. Prognostic value of preoperative enhanced computed tomography as a quantitative imaging biomarker in pancreatic cancer. World J Gastroenterol 2022; 28:2468-2481. [PMID: 35979266 PMCID: PMC9258279 DOI: 10.3748/wjg.v28.i22.2468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/31/2021] [Accepted: 05/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies with high mortality and short survival time. Computed tomography (CT) plays an important role in the diagnosis, staging and treatment of pancreatic tumour. Pancreatic cancer generally shows a low enhancement pattern compared with normal pancreatic tissue.
AIM To analyse whether preoperative enhanced CT could be used to predict postoperative overall survival in patients with PDAC.
METHODS Sixty-seven patients with PDAC undergoing pancreatic resection were enrolled retrospectively. All patients underwent preoperative unenhanced and enhanced CT examination, the CT values of which were measured. The ratio of the preoperative CT value increase from the nonenhancement phase to the portal venous phase between pancreatic tumour and normal pancreatic tissue was calculated. The cut-off value of ratios was obtained by the receiver operating characteristic (ROC) curve of the tumour relative enhancement ratio (TRER), according to which patients were divided into low- and high-enhancement groups. Univariate and multivariate analyses were performed using Cox regression based on TRER grouping. Finally, the correlation between TRER and clinicopathological characteristics was analysed.
RESULTS The area under the curve of the ROC curve was 0.768 (P < 0.05), and the cut-off value of the ROC curve was calculated as 0.7. TRER ≤ 0.7 was defined as the low-enhancement group, and TRER > 0.7 was defined as the high-enhancement group. According to the TRER grouping, the Kaplan-Meier survival curve analysis results showed that the median survival (10.0 mo) with TRER ≤ 0.7 was significantly shorter than that (22.0 mo) with TRER > 0.7 (P < 0.05). In the univariate and multivariate analyses, the prognosis of patients with TRER ≤ 0.7 was significantly worse than that of patients with TRER > 0.7 (P < 0.05). Our results demonstrated that patients in the low TRER group were more likely to have higher American Joint Committee on Cancer stage, tumour stage and lymph node stage (all P < 0.05), and TRER was significantly negatively correlated with tumour size (P < 0.05).
CONCLUSION TRER ≤ 0.7 in patients with PDAC may represent a tumour with higher clinical stage and result in a shorter overall survival.
Collapse
Affiliation(s)
- Jian-Feng Gao
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Yu Pan
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Xian-Chao Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Feng-Chun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Ding-Shen Qiu
- Department of Radiology, The Hospital of Changle, Fuzhou 350200, Fujian Province, China
| | - Jun-Jun Liu
- Department of Radiology, The Hospital of Changle, Fuzhou 350200, Fujian Province, China
| | - He-Guang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| |
Collapse
|
40
|
Quantitative MRI of Pancreatic Cystic Lesions: A New Diagnostic Approach. Healthcare (Basel) 2022; 10:healthcare10061039. [PMID: 35742090 PMCID: PMC9222599 DOI: 10.3390/healthcare10061039] [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: 04/29/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 02/01/2023] Open
Abstract
The commonly used magnetic resonance (MRI) criteria can be insufficient for discriminating mucinous from non-mucinous pancreatic cystic lesions (PCLs). The histological differences between PCLs’ fluid composition may be reflected in MRI images, but cannot be assessed by visual evaluation alone. We investigate whether additional MRI quantitative parameters such as signal intensity measurements (SIMs) and radiomics texture analysis (TA) can aid the differentiation between mucinous and non-mucinous PCLs. Fifty-nine PCLs (mucinous, n = 24; non-mucinous, n = 35) are retrospectively included. The SIMs were performed by two radiologists on T2 and diffusion-weighted images (T2WI and DWI) and apparent diffusion coefficient (ADC) maps. A total of 550 radiomic features were extracted from the T2WI and ADC maps of every lesion. The SIMs and TA features were compared between entities using univariate, receiver-operating, and multivariate analysis. The SIM analysis showed no statistically significant differences between the two groups (p = 0.69, 0.21–0.43, and 0.98 for T2, DWI, and ADC, respectively). Mucinous and non-mucinous PLCs were successfully discriminated by both T2-based (83.2–100% sensitivity and 69.3–96.2% specificity) and ADC-based (40–85% sensitivity and 60–96.67% specificity) radiomic features. SIMs cannot reliably discriminate between PCLs. Radiomics have the potential to augment the common MRI diagnosis of PLCs by providing quantitative and reproducible imaging features, but validation is required by further studies.
Collapse
|
41
|
Bekci T, Cakir IM, Aslan S. Differentiation of affected and nonaffected ovaries in ovarian torsion with magnetic resonance imaging texture analysis. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2022; 68:641-646. [PMID: 35584489 DOI: 10.1590/1806-9282.20211369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 01/03/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study aimed to evaluate the feasibility of texture analysis on T2-weighted axial images in differentiating affected and nonaffected ovaries in ovarian torsion. METHODS We included 22 torsioned ovaries and 19 healthy ovaries. All patients were surgically proven ovarian torsion cases. On T2-weighted axial images, ovarian borders were delineated by the consensus of two radiologists for magnetic resonance imaging-based texture analysis. Statistical differences between texture features of affected and nonaffected ovaries were assessed. RESULTS A total of 44 texture features were extracted from each ovary using LIFEx software. Of these, 17 features were significantly different between affected and nonaffected ovaries in ovarian torsion. NGLDM_Coarseness and NGLDM_Contrast, which are the neighborhood gray-level difference matrix parameters, had the largest area under the curve: 0.923. The best cutoff values for the NGLDM_Contrast and NGLDM_Coarseness were 0.45 and 0.01, respectively. With these cutoff levels, NGLDM_Contrast had the best accuracy (85.37%). CONCLUSION Magnetic resonance imaging-based texture analysis on axial T2-weighted images may help differentiate affected and nonaffected ovaries in ovarian torsion.
Collapse
Affiliation(s)
- Tumay Bekci
- Giresun University Faculty of Medicine, Department of Radiology - Giresun, Turkey
| | - Ismet Mirac Cakir
- Giresun University Faculty of Medicine, Department of Radiology - Giresun, Turkey
| | - Serdar Aslan
- Giresun University Faculty of Medicine, Department of Radiology - Giresun, Turkey
| |
Collapse
|
42
|
Yang Q, Mao Y, Xie H, Qin T, Mai Z, Cai Q, Wen H, Li Y, Zhang R, Liu L. Identifying Outcomes of Patients With Advanced Pancreatic Adenocarcinoma and RECIST Stable Disease Using Radiomics Analysis. JCO Precis Oncol 2022; 6:e2100362. [PMID: 35319966 PMCID: PMC8966975 DOI: 10.1200/po.21.00362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Few studies have explored the biomarkers for predicting the heterogeneous outcomes of patients with advanced pancreatic adenocarcinoma showing stable disease (SD) on the initial postchemotherapy computed tomography. We aimed to devise a radiomics signature (RS) to predict these outcomes for further risk stratification.
Collapse
Affiliation(s)
- Qiuxia Yang
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yize Mao
- Department of Pancreatic-Biliary Surgical Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hui Xie
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Tao Qin
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhijun Mai
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qian Cai
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hailin Wen
- Cancer Hospital Chinese Academy of Medical Science, Shenzhen Center, Shenzhen, China
| | - Yong Li
- Department of Medical Imaging Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rong Zhang
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lizhi Liu
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
43
|
Rossi G, Altabella L, Simoni N, Benetti G, Rossi R, Venezia M, Paiella S, Malleo G, Salvia R, Guariglia S, Bassi C, Cavedon C, Mazzarotto R. Computed tomography-based radiomic to predict resectability in locally advanced pancreatic cancer treated with chemotherapy and radiotherapy. World J Gastrointest Oncol 2022; 14:703-715. [PMID: 35321278 PMCID: PMC8919018 DOI: 10.4251/wjgo.v14.i3.703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/06/2021] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Surgical resection after neoadjuvant treatment is the main driver for improved survival in locally advanced pancreatic cancer (LAPC). However, the diagnostic performance of computed tomography (CT) imaging to evaluate the residual tumour burden at restaging after neoadjuvant therapy is low due to the difficulty in distinguishing neoplastic tissue from fibrous scar or inflammation. In this context, radiomics has gained popularity over conventional imaging as a complementary clinical tool capable of providing additional, unprecedented information regarding the intratumor heterogeneity and the residual neoplastic tissue, potentially serving in the therapeutic decision-making process. AIM To assess the capability of radiomic features to predict surgical resection in LAPC treated with neoadjuvant chemotherapy and radiotherapy. METHODS Patients with LAPC treated with intensive chemotherapy followed by ablative radiation therapy were retrospectively reviewed. One thousand six hundred and fifty-five radiomic features were extracted from planning CT inside the gross tumour volume. Both extracted features and clinical data contribute to create and validate the predictive model of resectability status. Patients were repeatedly divided into training and validation sets. The discriminating performance of each model, obtained applying a LASSO regression analysis, was assessed with the area under the receiver operating characteristic curve (AUC). The validated model was applied to the entire dataset to obtain the most significant features. RESULTS Seventy-one patients were included in the analysis. Median age was 65 years and 57.8% of patients were male. All patients underwent induction chemotherapy followed by ablative radiotherapy, and 19 (26.8%) ultimately received surgical resection. After the first step of variable selections, a predictive model of resectability was developed with a median AUC for training and validation sets of 0.862 (95%CI: 0.792-0.921) and 0.853 (95%CI: 0.706-0.960), respectively. The validated model was applied to the entire dataset and 4 features were selected to build the model with predictive performance as measured using AUC of 0.944 (95%CI: 0.892-0.996). CONCLUSION The present radiomic model could help predict resectability in LAPC after neoadjuvant chemotherapy and radiotherapy, potentially integrating clinical and morphological parameters in predicting surgical resection.
Collapse
Affiliation(s)
- Gabriella Rossi
- Department of Radiation Oncology, University of Verona Hospital Trust, Verona 37126, Italy
| | - Luisa Altabella
- Department of Medical Physics, University of Verona Hospital Trust, Verona 37126, Italy
| | - Nicola Simoni
- Department of Radiation Oncology, University of Verona Hospital Trust, Verona 37126, Italy
| | - Giulio Benetti
- Department of Medical Physics, University of Verona Hospital Trust, Verona 37126, Italy
| | - Roberto Rossi
- Department of Radiation Oncology, University of Verona Hospital Trust, Verona 37126, Italy
| | - Martina Venezia
- Department of Radiation Oncology, University of Verona Hospital Trust, Verona 37126, Italy
| | - Salvatore Paiella
- Department of General and Pancreatic Surgery, Pancreas Institute, University of Verona Hospital Trust, Verona 37126, Italy
| | - Giuseppe Malleo
- Department of General and Pancreatic Surgery, Pancreas Institute, University of Verona Hospital Trust, Verona 37126, Italy
| | - Roberto Salvia
- Department of General and Pancreatic Surgery, Pancreas Institute, University of Verona Hospital Trust, Verona 37126, Italy
| | - Stefania Guariglia
- Department of Medical Physics, University of Verona Hospital Trust, Verona 37126, Italy
| | - Claudio Bassi
- Department of General and Pancreatic Surgery, Pancreas Institute, University of Verona Hospital Trust, Verona 37126, Italy
| | - Carlo Cavedon
- Department of Medical Physics, University of Verona Hospital Trust, Verona 37126, Italy
| | - Renzo Mazzarotto
- Department of Radiation Oncology, University of Verona Hospital Trust, Verona 37126, Italy
| |
Collapse
|
44
|
Radiofrequency ablation of hepatocellular carcinoma: CT texture analysis of the ablated area to predict local recurrence. Eur J Radiol 2022; 150:110250. [DOI: 10.1016/j.ejrad.2022.110250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 11/22/2022]
|
45
|
Zhang X, Zhang Y, Zhang G, Qiu X, Tan W, Yin X, Liao L. Deep Learning With Radiomics for Disease Diagnosis and Treatment: Challenges and Potential. Front Oncol 2022; 12:773840. [PMID: 35251962 PMCID: PMC8891653 DOI: 10.3389/fonc.2022.773840] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
The high-throughput extraction of quantitative imaging features from medical images for the purpose of radiomic analysis, i.e., radiomics in a broad sense, is a rapidly developing and emerging research field that has been attracting increasing interest, particularly in multimodality and multi-omics studies. In this context, the quantitative analysis of multidimensional data plays an essential role in assessing the spatio-temporal characteristics of different tissues and organs and their microenvironment. Herein, recent developments in this method, including manually defined features, data acquisition and preprocessing, lesion segmentation, feature extraction, feature selection and dimension reduction, statistical analysis, and model construction, are reviewed. In addition, deep learning-based techniques for automatic segmentation and radiomic analysis are being analyzed to address limitations such as rigorous workflow, manual/semi-automatic lesion annotation, and inadequate feature criteria, and multicenter validation. Furthermore, a summary of the current state-of-the-art applications of this technology in disease diagnosis, treatment response, and prognosis prediction from the perspective of radiology images, multimodality images, histopathology images, and three-dimensional dose distribution data, particularly in oncology, is presented. The potential and value of radiomics in diagnostic and therapeutic strategies are also further analyzed, and for the first time, the advances and challenges associated with dosiomics in radiotherapy are summarized, highlighting the latest progress in radiomics. Finally, a robust framework for radiomic analysis is presented and challenges and recommendations for future development are discussed, including but not limited to the factors that affect model stability (medical big data and multitype data and expert knowledge in medical), limitations of data-driven processes (reproducibility and interpretability of studies, different treatment alternatives for various institutions, and prospective researches and clinical trials), and thoughts on future directions (the capability to achieve clinical applications and open platform for radiomics analysis).
Collapse
Affiliation(s)
- Xingping Zhang
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, China
| | - Yanchun Zhang
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
- Department of New Networks, Peng Cheng Laboratory, Shenzhen, China
| | - Guijuan Zhang
- Department of Respiratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xingting Qiu
- Department of Radiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Wenjun Tan
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Shenyang, China
| | - Xiaoxia Yin
- Institute of Advanced Cyberspace Technology, Guangzhou University, Guangzhou, China
| | - Liefa Liao
- School of Information Engineering, Jiangxi University of Science and Technology, Ganzhou, China
| |
Collapse
|
46
|
Ma X, Wang YR, Zhuo LY, Yin XP, Ren JL, Li CY, Xing LH, Zheng TT. Retrospective Analysis of the Value of Enhanced CT Radiomics Analysis in the Differential Diagnosis Between Pancreatic Cancer and Chronic Pancreatitis. Int J Gen Med 2022; 15:233-241. [PMID: 35023961 PMCID: PMC8747707 DOI: 10.2147/ijgm.s337455] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose To investigate the feasibility of enhanced computed tomography (CT) radiomics analysis to differentiate between pancreatic cancer (PC) and chronic pancreatitis. Methods and materials The CT images of 151 PCs and 24 chronic pancreatitis were retrospectively analyzed in the three-dimensional regions of interest on arterial phase (AP) and venous phase (VP) and segmented by MITK software. A multivariable logistic regression model was established based on the selected radiomics features. The radiomics score was calculated, and the nomogram was established. The discrimination of each model was analyzed by the receiver operating characteristic curve (ROC). Decision curve analysis (DCA) was used to evaluate clinical utility. The precision recall curve (PRC) was used to evaluate whether the model is affected by data imbalance. The Delong test was adopted to compare the diagnostic efficiency of each model. Results Significant differences were observed in the distribution of gender (P = 0.034), carbohydrate antigen 19-9 (P < 0.001), and carcinoembryonic antigen (P < 0.001) in patients with PC and chronic pancreatitis. The area under the ROC curve (AUC) value of AP multivariate regression model, VP multivariate regression model, AP combined with VP features model (Radiomics), clinical feature model, and radiomics combined with clinical feature model (COMB) was 0.905, 0.941, 0.941, 0.822, and 0.980, respectively. The sensitivity and specificity of the COMB model were 0.947 and 0.917, respectively. The results of DCA showed that the COMB model exhibited net clinical benefits and PRC shows that COMB model have good precision and recall (sensitivity). Conclusion The COMB model could be a potential tool to distinguish PC from chronic pancreatitis and aid in clinical decisions.
Collapse
Affiliation(s)
- Xi Ma
- CT/MRI Room, Affiliated Hospital of Hebei University, Baoding, Hebei Province, 071000, People's Republic of China
| | - Yu-Rui Wang
- Department of Computed Tomography, Tangshan Gongren Hospital, Tangshan, Hebei Province, 063000, People's Republic of China
| | - Li-Yong Zhuo
- CT/MRI Room, Affiliated Hospital of Hebei University, Baoding, Hebei Province, 071000, People's Republic of China
| | - Xiao-Ping Yin
- CT/MRI Room, Affiliated Hospital of Hebei University, Baoding, Hebei Province, 071000, People's Republic of China
| | - Jia-Liang Ren
- GE Healthcare[Shanghai] Co Ltd, Shanghai, 210000, People's Republic of China
| | - Cai-Ying Li
- Department of Radiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, People's Republic of China
| | - Li-Hong Xing
- CT/MRI Room, Affiliated Hospital of Hebei University, Baoding, Hebei Province, 071000, People's Republic of China
| | - Tong-Tong Zheng
- Department of Radiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, People's Republic of China
| |
Collapse
|
47
|
Chen HY, Deng XY, Pan Y, Chen JY, Liu YY, Chen WJ, Yang H, Zheng Y, Yang YB, Liu C, Shao GL, Yu RS. Pancreatic Serous Cystic Neoplasms and Mucinous Cystic Neoplasms: Differential Diagnosis by Combining Imaging Features and Enhanced CT Texture Analysis. Front Oncol 2022; 11:745001. [PMID: 35004272 PMCID: PMC8733460 DOI: 10.3389/fonc.2021.745001] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/29/2021] [Indexed: 12/25/2022] Open
Abstract
Objective To establish a diagnostic model by combining imaging features with enhanced CT texture analysis to differentiate pancreatic serous cystadenomas (SCNs) from pancreatic mucinous cystadenomas (MCNs). Materials and Methods Fifty-seven and 43 patients with pathology-confirmed SCNs and MCNs, respectively, from one center were analyzed and divided into a training cohort (n = 72) and an internal validation cohort (n = 28). An external validation cohort (n = 28) from another center was allocated. Demographic and radiological information were collected. The least absolute shrinkage and selection operator (LASSO) and recursive feature elimination linear support vector machine (RFE_LinearSVC) were implemented to select significant features. Multivariable logistic regression algorithms were conducted for model construction. Receiver operating characteristic (ROC) curves for the models were evaluated, and their prediction efficiency was quantified by the area under the curve (AUC), 95% confidence interval (95% CI), sensitivity and specificity. Results Following multivariable logistic regression analysis, the AUC was 0.932 and 0.887, the sensitivity was 87.5% and 90%, and the specificity was 82.4% and 84.6% with the training and validation cohorts, respectively, for the model combining radiological features and CT texture features. For the model based on radiological features alone, the AUC was 0.84 and 0.91, the sensitivity was 75% and 66.7%, and the specificity was 82.4% and 77% with the training and validation cohorts, respectively. Conclusion This study showed that a logistic model combining radiological features and CT texture features is more effective in distinguishing SCNs from MCNs of the pancreas than a model based on radiological features alone.
Collapse
Affiliation(s)
- Hai-Yan Chen
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institue of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Xue-Ying Deng
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institue of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Yao Pan
- Department of Radiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie-Yu Chen
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institue of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Yun-Ying Liu
- Institue of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China.,Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Wu-Jie Chen
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institue of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Hong Yang
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institue of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Yao Zheng
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institue of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Yong-Bo Yang
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institue of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Cheng Liu
- Research Institute of Artificial Intelligence in Healthcare, Hangzhou YITU Healthcare Technology Co. Ltd., Hangzhou, China
| | - Guo-Liang Shao
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institue of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China.,Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Ri-Sheng Yu
- Department of Radiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
48
|
Non-contrast-enhanced CT texture analysis of primary and metastatic pancreatic ductal adenocarcinomas: value in assessment of histopathological grade and differences between primary and metastatic lesions. Abdom Radiol (NY) 2022; 47:4151-4159. [PMID: 36104481 PMCID: PMC9626421 DOI: 10.1007/s00261-022-03646-7] [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: 04/08/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE To evaluate the utility of non-contrast-enhanced CT texture analysis (CTTA) for predicting the histopathological differentiation of pancreatic ductal adenocarcinomas (PDAC) and to compare non-contrast-enhanced CTTA texture features between primary PDAC and hepatic metastases of PDAC. METHODS This retrospective study included 120 patients with histopathologically confirmed PDAC. Sixty-five patients underwent CT-guided biopsy of primary PDAC, while 55 patients underwent CT-guided biopsy of hepatic PDAC metastasis. All lesions were segmented in non-contrast-enhanced CT scans for CTTA based on histogram analysis, co-occurrence matrix, and run-length matrix. Statistical analysis was conducted for 372 texture features using Mann-Whitney U test, Bonferroni-Holm correction, and receiver operating characteristic (ROC) analysis. A p value < 0.05 was considered statistically significant. RESULTS Three features were identified that differed significantly between histopathological G2 and G3 primary tumors. Of these, "low gray-level zone emphasis" yielded the largest AUC (0.87 ± 0.04), reaching a sensitivity and specificity of 0.76 and 0.83, respectively, when a cut-off value of 0.482 was applied. Fifty-four features differed significantly between primary and hepatic metastatic PDAC. CONCLUSION Non-contrast-enhanced CTTA of PDAC identified differences in texture features between primary G2 and G3 tumors that could be used for non-invasive tumor assessment. Extensive differences between the features of primary and metastatic PDAC on CTTA suggest differences in tumor microenvironment.
Collapse
|
49
|
Park HJ, Qin L, Bakouny Z, Krajewski KM, Van Allen EM, Choueiri TK, Shinagare AB. OUP accepted manuscript. Oncologist 2022; 27:389-397. [PMID: 35348767 PMCID: PMC9074990 DOI: 10.1093/oncolo/oyac034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 01/07/2022] [Indexed: 11/15/2022] Open
Abstract
Background Materials and Methods Results Conclusion
Collapse
Affiliation(s)
- Hyo Jung Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Lei Qin
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Katherine M Krajewski
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Atul B Shinagare
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Corresponding author: Atul B. Shinagare, Department of Radiology, Brigham and Womens Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA. Tel.: +1 6176322988; Fax: +1 6175828574;
| |
Collapse
|
50
|
Li X, Wan Y, Lou J, Xu L, Shi A, Yang L, Fan Y, Yang J, Huang J, Wu Y, Niu T. Preoperative recurrence prediction in pancreatic ductal adenocarcinoma after radical resection using radiomics of diagnostic computed tomography. EClinicalMedicine 2022; 43:101215. [PMID: 34927034 PMCID: PMC8649647 DOI: 10.1016/j.eclinm.2021.101215] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The high recurrence rate after radical resection of pancreatic ductal adenocarcinoma (PDAC) leads to its poor prognosis. We aimed to develop a model to preoperatively predict the risk of recurrence based on computed tomography (CT) radiomics and multiple clinical parameters. METHODS Datasets were retrospectively collected and analysed of 220 PDAC patients who underwent contrast-enhanced computed tomography (CE-CT) and received radical resection at 3 institutions in China between 2013 and 2017, with 153 from one institution as a training set, the remaining 67 as a validation set. For each patient, CT radiomics features were extracted from intratumoral and peritumoral regions to establish intratumoral, peritumoral and combined radiomics models using artificial neural network (ANN) algorithm. By incorporating clinical factors, radiomics-clinical nomograms were finally built by multivariable logistic regression analysis to predict 1- and 2-year recurrence risk. FINDINGS The developed radiomics model integrating intratumoral and peritumoral radiomics features was superior to the conventionally constructed model merely using intratumoral radiomics features. Further, radiomics-clinical nomograms outperformed other models in predicting 1-year recurrence with an area under the receiver operating characteristic curve (AUROC) of 0.916 (95%CI, 0.860-0.955) in the training set and 0.764 (95%CI, 0.644-0.859) in the validation set, and 2-year recurrence with an AUROC of 0.872 (95%CI: 0.809-0.921) in the training set and 0.773 (95%CI, 0.654-0.866) in the validation set. INTERPRETATION This study has developed and externally validated a radiomics-clinical nomogram integrating intra- and peritumoral CT radiomics signature as well as clinical factors to predict the recurrence risk of PDAC after radical resection, which will facilitate optimized and individualized treatment strategies. FUNDING This work was supported by the National Key R&D Program of China [grant number: 2018YFE0114800], the General Program of National Natural Science Foundation of China [grant number: 81772562, 2017; 81871351, 2018], the Fundamental Research Funds for the Central Universities [grant number: 2021FZZX005-08], and Zhejiang Provincial Key Projects of Technology Research [grant number: WKJ-ZJ-2033].
Collapse
Affiliation(s)
- Xiawei Li
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yidong Wan
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianyao Lou
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lei Xu
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Aiguang Shi
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Litao Yang
- Department of Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Yiqun Fan
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Jing Yang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junjie Huang
- Department of Surgery, Changxing People's Hospital, Huzhou, Zhejiang, China
| | - Yulian Wu
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tianye Niu
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|