1
|
Shelly M, Sivakumari S. Enhancing pancreatic cancer diagnostics: Ensemble-based model for automated urine biomarker classification. Comput Biol Med 2025; 189:109997. [PMID: 40068492 DOI: 10.1016/j.compbiomed.2025.109997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
This research addresses the critical challenge of early detection in pancreatic ductal adenocarcinoma (PDAC) by exploring urinary biomarkers and integrating artificial intelligence (AI) models. The study emphasizes the significance of liquid biopsy, particularly in urine, as a noninvasive approach for real-time monitoring of PDAC. Traditional diagnostic methods face limitations, necessitating the exploration of novel biomarkers and AI applications. The research employs two distinct methodologies: Method 1, utilizing a Bagged Ensemble of decision trees, and Method 2, employing an optimized decision tree model. Comprehensive evaluations, including accuracy, confusion matrices, and ROC curves, reveal the effectiveness of both methods in PDAC diagnostics. The comparison highlights Method 1's marginally superior performance in training, while Method 2 excels in distinguishing between benign and PDAC cases during testing. The research underscores the potential of AI-driven urinary biomarker classification as a robust diagnostic tool for improved outcomes in pancreatic cancer The evaluation results against state-of-the-art models, further emphasize the superiority of the proposed ensemble-based methods.
Collapse
Affiliation(s)
- Minnuja Shelly
- Computer Science and Engineering Department, Avinashilingam Institute for Home Science and Education for Women, School of Engineering, Coimbatore, India.
| | - S Sivakumari
- Computer Science and Engineering Department, Avinashilingam Institute for Home Science and Education for Women, School of Engineering, Coimbatore, India.
| |
Collapse
|
2
|
Zhang X, Gao A, Ma L, Yu N. Integrating intratumoral and peritumoral radiomics with clinical risk factors for prognostic prediction in pancreatic ductal adenocarcinoma patients undergoing combined chemotherapy and HIFU ablation. Int J Hyperthermia 2024; 41:2410342. [PMID: 39353582 DOI: 10.1080/02656736.2024.2410342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
OBJECTIVE A radiomics nomogram will be created utilizing MRI data from intratumoral and peritumoral areas to forecast survival outcomes in patients who have had treatment for pancreatic ductal adenocarcinoma (PDAC). METHODS A total of 87 individuals diagnosed with PDAC were included in the study, with 60 patients in the training cohort and 27 patients in the validation cohort. A grand total of 2395 radiomics characteristics were extracted from the tumor region and the peritumoral region. The least absolute shrinkage and selection operator (LASSO) method was used to select features and create a radiomics score, also known as the Rad-score. A multivariate regression analysis was then conducted to build the radiomics nomogram. The evaluation of the nomogram included discrimination, calibration, and clinical utility assessments. RESULTS Based on the conclusions derived from the multivariate Cox model, Rad-Score, jaundice, and tumor size were identified as independent risk factors for overall survival (OS). The inclusion of the Rad-score in the radiomics nomogram led to improved accuracy in predicting survival compared to the clinical model. Patients were categorized into high-risk and low-risk groups based on their Rad-Score. Kaplan-Meier analysis revealed a statistically significant difference between the two groups (p < 0.05). Furthermore, the radiomics nomogram demonstrated excellent ability to differentiate, calibrate, and provide clinical utility in both the training and validation groups. CONCLUSIONS The MRI-based intratumoral and peritumoral radiomics nomogram, integrating the Rad-score and clinical data, provided better prognostic prediction for PDAC patients after HIFU treatment, which may hold great potential for guiding personalized care for these patients.
Collapse
Affiliation(s)
- Xuehui Zhang
- Department of Ultrasound, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aixin Gao
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Leiyuan Ma
- Department of Ultrasound, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ning Yu
- Department of Ultrasound, the Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
3
|
Jiang L, Ye Y, Feng Z, Liu W, Cao Y, Zhao X, Zhu X, Zhang H. Stereotactic body radiation therapy for the primary tumor and oligometastases versus the primary tumor alone in patients with metastatic pancreatic cancer. Radiat Oncol 2024; 19:111. [PMID: 39160547 PMCID: PMC11334573 DOI: 10.1186/s13014-024-02493-8] [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: 01/10/2024] [Accepted: 07/19/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Local therapies may benefit patients with oligometastatic cancer. However, there were limited data about pancreatic cancer. Here, we compared the efficacy and safety of stereotactic body radiation therapy (SBRT) to the primary tumor and all oligometastases with SBRT to the primary tumor alone in patients with metastatic pancreatic cancer. METHODS A retrospective review of patients with synchronous oligometastatic pancreatic cancer (up to 5 lesions) receiving SBRT to all lesions (including all oligometastases and the primary tumor) were performed. Another comparable group of patients with similar baseline characteristics, including metastatic burden, SBRT doses, and chemotherapy regimens, receiving SBRT to the primary tumor alone were identified. The primary endpoint was overall survival (OS). The secondary endpoints were progression frees survival (PFS), polyprogression free survival (PPFS) and adverse events. RESULTS There were 59 and 158 patients receiving SBRT to all lesions and to the primary tumor alone. The median OS of patients with SBRT to all lesions and the primary tumor alone was 10.9 months (95% CI 10.2-11.6 months) and 9.3 months (95% CI 8.8-9.8 months) (P < 0.001). The median PFS of two groups was 6.5 months (95% CI 5.6-7.4 months) and 4.1 months (95% CI 3.8-4.4 months) (P < 0.001). The median PPFS of two groups was 9.8 months (95% CI 8.9-10.7 months) and 7.8 months (95% CI 7.2-8.4 months) (P < 0.001). Additionally, 14 (23.7%) and 32 (20.2%) patients in two groups had grade 3 or 4 treatment-related toxicity. CONCLUSIONS SBRT to all oligometastases and the primary tumor in patients with pancreatic cancer may improve survival, which needs prospective verification.
Collapse
Affiliation(s)
- Lingong Jiang
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yusheng Ye
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Zhiru Feng
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Wenyu Liu
- Department of Hepatobiliary and Pancreatic Surgery, Changhai Hospital affiliated to Naval Medical University, Shanghai, China
| | - Yangsen Cao
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xianzhi Zhao
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiaofei Zhu
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Huojun Zhang
- Department of Radiation Oncology, Changhai Hospital affiliated to Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
4
|
Zhang Z, Yu G, Eresen A, Chen Z, Yu Z, Yaghmai V, Zhang Z. Dendritic cell vaccination combined with irreversible electroporation for treating pancreatic cancer-a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:77. [PMID: 39118942 PMCID: PMC11304422 DOI: 10.21037/atm-23-1882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/25/2024] [Indexed: 08/10/2024]
Abstract
Background and Objective Pancreatic ductal adenocarcinoma (PDAC) is 3rd most lethal cancer in the USA leading to a median survival of six months and less than 5% 5-year overall survival (OS). As the only potentially curative treatment, surgical resection is not suitable for up to 90% of the patients with PDAC due to late diagnosis. Highly fibrotic PDAC with an immunosuppressive tumor microenvironment restricts cytotoxic T lymphocyte (CTL) infiltration and functions causing limited success with systemic therapies like dendritic cell (DC)-based immunotherapy. In this study, we investigated the potential benefits of irreversible electroporation (IRE) ablation therapy in combination with DC vaccine therapy against PDAC. Methods We performed a literature search to identify studies focused on DC vaccine therapy and IRE ablation to boost therapeutic response against PDAC indexed in PubMed, Web of Science, and Scopus until February 20th, 2023. Key Content and Findings IRE ablation destructs tumor structure while preserving extracellular matrix and blood vessels facilitating local inflammation. The studies demonstrated IRE ablation reduces tumor fibrosis and promotes CTL tumor infiltration to PDAC tumors in addition to boosting immune response in rodent models. The administration of the DC vaccine following IRE ablation synergistically enhances therapeutic response and extends OS rates compared to the use of DC vaccination or IRE alone. Moreover, the implementation of data-driven approaches further allows dynamic and longitudinal monitoring of therapeutic response and OS following IRE plus DC vaccine immunoablation. Conclusions The combination of IRE ablation and DC vaccine immunotherapy is a potent strategy to enhance the therapeutic outcomes in patients with PDAC.
Collapse
Affiliation(s)
- Zigeng Zhang
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
| | - Guangbo Yu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Aydin Eresen
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
| | - Zhilin Chen
- Department of Human Biology and Business Administration, University of Southern California, Los Angeles, CA, USA
| | - Zeyang Yu
- Information School, University of Washington, Seattle, WA, USA
| | - Vahid Yaghmai
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Zhuoli Zhang
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
5
|
Nopour R. Establishment of prediction model for mortality risk of pancreatic cancer: a retrospective study. BMC Med Inform Decis Mak 2024; 24:181. [PMID: 38937795 PMCID: PMC11210158 DOI: 10.1186/s12911-024-02590-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/25/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND AND AIM Pancreatic cancer possesses a high prevalence and mortality rate among other cancers. Despite the low survival rate of this cancer type, the early prediction of this disease has a crucial role in decreasing the mortality rate and improving the prognosis. So, this study. MATERIALS AND METHODS In this retrospective study, we used 654 alive and dead PC cases to establish the prediction model for PC. The six chosen machine learning algorithms and prognostic factors were utilized to build the prediction models. The importance of the predictive factors was assessed using the relative importance of a high-performing algorithm. RESULTS The XG-Boost with AU-ROC of 0.933 (95% CI= [0.906-0.958]) and AU-ROC of 0.836 (95% CI= [0.789-0.865] in internal and external validation modes were considered as the best-performing model for predicting the mortality risk of PC. The factors, including tumor size, smoking, and chemotherapy, were considered the most influential for prediction. CONCLUSION The XG-Boost gained more performance efficiency in predicting the mortality risk of PC patients, so this model can promote the clinical solutions that doctors can achieve in healthcare environments to decrease the mortality risk of these patients.
Collapse
Affiliation(s)
- Raoof Nopour
- Department of Health Information Management, Student Research Committee, School of Health Management and Information Sciences Branch, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Liao H, Yuan J, Liu C, Zhang J, Yang Y, Liang H, Jiang S, Chen S, Li Y, Liu Y. Feasibility and effectiveness of automatic deep learning network and radiomics models for differentiating tumor stroma ratio in pancreatic ductal adenocarcinoma. Insights Imaging 2023; 14:223. [PMID: 38129708 PMCID: PMC10739634 DOI: 10.1186/s13244-023-01553-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/28/2023] [Indexed: 12/23/2023] Open
Abstract
OBJECTIVE This study aims to compare the feasibility and effectiveness of automatic deep learning network and radiomics models in differentiating low tumor stroma ratio (TSR) from high TSR in pancreatic ductal adenocarcinoma (PDAC). METHODS A retrospective analysis was conducted on a total of 207 PDAC patients from three centers (training cohort: n = 160; test cohort: n = 47). TSR was assessed on hematoxylin and eosin-stained specimens by experienced pathologists and divided as low TSR and high TSR. Deep learning and radiomics models were developed including ShuffulNetV2, Xception, MobileNetV3, ResNet18, support vector machine (SVM), k-nearest neighbor (KNN), random forest (RF), and logistic regression (LR). Additionally, the clinical models were constructed through univariate and multivariate logistic regression. Kaplan-Meier survival analysis and log-rank tests were conducted to compare the overall survival time between different TSR groups. RESULTS To differentiate low TSR from high TSR, the deep learning models based on ShuffulNetV2, Xception, MobileNetV3, and ResNet18 achieved AUCs of 0.846, 0.924, 0.930, and 0.941, respectively, outperforming the radiomics models based on SVM, KNN, RF, and LR with AUCs of 0.739, 0.717, 0.763, and 0.756, respectively. Resnet 18 achieved the best predictive performance. The clinical model based on T stage alone performed worse than deep learning models and radiomics models. The survival analysis based on 142 of the 207 patients demonstrated that patients with low TSR had longer overall survival. CONCLUSIONS Deep learning models demonstrate feasibility and superiority over radiomics in differentiating TSR in PDAC. The tumor stroma ratio in the PDAC microenvironment plays a significant role in determining prognosis. CRITICAL RELEVANCE STATEMENT The objective was to compare the feasibility and effectiveness of automatic deep learning networks and radiomics models in identifying the tumor-stroma ratio in pancreatic ductal adenocarcinoma. Our findings demonstrate deep learning models exhibited superior performance compared to traditional radiomics models. KEY POINTS • Deep learning demonstrates better performance than radiomics in differentiating tumor-stroma ratio in pancreatic ductal adenocarcinoma. • The tumor-stroma ratio in the pancreatic ductal adenocarcinoma microenvironment plays a protective role in prognosis. • Preoperative prediction of tumor-stroma ratio contributes to clinical decision-making and guiding precise medicine.
Collapse
Affiliation(s)
- Hongfan Liao
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiang Yuan
- College of Computer and Information Science, Southwest University, Chongqing, 400715, China
| | - Chunhua Liu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jiao Zhang
- Department of Radiology, the Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaying Yang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Hongwei Liang
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Song Jiang
- Department of Radiology, Chongqing Ping An Medical Imaging Diagnosis Center, Chongqing, China
| | - Shanxiong Chen
- College of Computer and Information Science, Southwest University, Chongqing, 400715, China.
| | - Yongmei Li
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Yanbing Liu
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
7
|
Mascharak S, Guo JL, Foster DS, Khan A, Davitt MF, Nguyen AT, Burcham AR, Chinta MS, Guardino NJ, Griffin M, Lopez DM, Miller E, Januszyk M, Raghavan SS, Longacre TA, Delitto DJ, Norton JA, Longaker MT. Desmoplastic stromal signatures predict patient outcomes in pancreatic ductal adenocarcinoma. Cell Rep Med 2023; 4:101248. [PMID: 37865092 PMCID: PMC10694604 DOI: 10.1016/j.xcrm.2023.101248] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/01/2023] [Accepted: 09/26/2023] [Indexed: 10/23/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is projected to become the second leading cause of cancer-related death. Hallmarks include desmoplasia with variable extracellular matrix (ECM) architecture and a complex microenvironment with spatially defined tumor, stromal, and immune populations. Nevertheless, the role of desmoplastic spatial organization in patient/tumor variability remains underexplored, which we elucidate using two technologies. First, we quantify ECM patterning in 437 patients, revealing architectures associated with disease-free and overall survival. Second, we spatially profile the cellular milieu of 78 specimens using codetection by indexing, identifying an axis of pro-inflammatory cell interactions predictive of poorer outcomes. We discover that clinical characteristics, including neoadjuvant chemotherapy status, tumor stage, and ECM architecture, correlate with differential stromal-immune organization, including fibroblast subtypes with distinct niches. Lastly, we define unified signatures that predict survival with areas under the receiver operating characteristic curve (AUCs) of 0.872-0.903, differentiating survivorship by 655 days. Overall, our findings establish matrix ultrastructural and cellular organizations of fibrosis linked to poorer outcomes.
Collapse
Affiliation(s)
- Shamik Mascharak
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jason L Guo
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Deshka S Foster
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anum Khan
- Cell Sciences Imaging Facility, Stanford University, Stanford, CA 94305, USA
| | - Michael F Davitt
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan T Nguyen
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Austin R Burcham
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Malini S Chinta
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nicholas J Guardino
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle Griffin
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David M Lopez
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elisabeth Miller
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Michael Januszyk
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shyam S Raghavan
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Teri A Longacre
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daniel J Delitto
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jeffrey A Norton
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Michael T Longaker
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
8
|
Ramaekers M, Viviers CGA, Janssen BV, Hellström TAE, Ewals L, van der Wulp K, Nederend J, Jacobs I, Pluyter JR, Mavroeidis D, van der Sommen F, Besselink MG, Luyer MDP. Computer-Aided Detection for Pancreatic Cancer Diagnosis: Radiological Challenges and Future Directions. J Clin Med 2023; 12:4209. [PMID: 37445243 DOI: 10.3390/jcm12134209] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Radiological imaging plays a crucial role in the detection and treatment of pancreatic ductal adenocarcinoma (PDAC). However, there are several challenges associated with the use of these techniques in daily clinical practice. Determination of the presence or absence of cancer using radiological imaging is difficult and requires specific expertise, especially after neoadjuvant therapy. Early detection and characterization of tumors would potentially increase the number of patients who are eligible for curative treatment. Over the last decades, artificial intelligence (AI)-based computer-aided detection (CAD) has rapidly evolved as a means for improving the radiological detection of cancer and the assessment of the extent of disease. Although the results of AI applications seem promising, widespread adoption in clinical practice has not taken place. This narrative review provides an overview of current radiological CAD systems in pancreatic cancer, highlights challenges that are pertinent to clinical practice, and discusses potential solutions for these challenges.
Collapse
Affiliation(s)
- Mark Ramaekers
- Department of Surgery, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands
| | - Christiaan G A Viviers
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Boris V Janssen
- Department of Surgery, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Terese A E Hellström
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Lotte Ewals
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands
| | - Kasper van der Wulp
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands
| | - Joost Nederend
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands
| | - Igor Jacobs
- Department of Hospital Services and Informatics, Philips Research, 5656 AE Eindhoven, The Netherlands
| | - Jon R Pluyter
- Department of Experience Design, Philips Design, 5656 AE Eindhoven, The Netherlands
| | - Dimitrios Mavroeidis
- Department of Data Science, Philips Research, 5656 AE Eindhoven, The Netherlands
| | - Fons van der Sommen
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Marc G Besselink
- Department of Surgery, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Misha D P Luyer
- Department of Surgery, Catharina Cancer Institute, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, The Netherlands
| |
Collapse
|
9
|
Dinesh MG, Bacanin N, Askar SS, Abouhawwash M. Diagnostic ability of deep learning in detection of pancreatic tumour. Sci Rep 2023; 13:9725. [PMID: 37322046 PMCID: PMC10272117 DOI: 10.1038/s41598-023-36886-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023] Open
Abstract
Pancreatic cancer is associated with higher mortality rates due to insufficient diagnosis techniques, often diagnosed at an advanced stage when effective treatment is no longer possible. Therefore, automated systems that can detect cancer early are crucial to improve diagnosis and treatment outcomes. In the medical field, several algorithms have been put into use. Valid and interpretable data are essential for effective diagnosis and therapy. There is much room for cutting-edge computer systems to develop. The main objective of this research is to predict pancreatic cancer early using deep learning and metaheuristic techniques. This research aims to create a deep learning and metaheuristic techniques-based system to predict pancreatic cancer early by analyzing medical imaging data, mainly CT scans, and identifying vital features and cancerous growths in the pancreas using Convolutional Neural Network (CNN) and YOLO model-based CNN (YCNN) models. Once diagnosed, the disease cannot be effectively treated, and its progression is unpredictable. That's why there's been a push in recent years to implement fully automated systems that can sense cancer at a prior stage and improve diagnosis and treatment. The paper aims to evaluate the effectiveness of the novel YCNN approach compared to other modern methods in predicting pancreatic cancer. To predict the vital features from the CT scan and the proportion of cancer feasts in the pancreas using the threshold parameters booked as markers. This paper employs a deep learning approach called a Convolutional Neural network (CNN) model to predict pancreatic cancer images. In addition, we use the YOLO model-based CNN (YCNN) to aid in the categorization process. Both biomarkers and CT image dataset is used for testing. The YCNN method was shown to perform well by a cent percent of accuracy compared to other modern techniques in a thorough review of comparative findings.
Collapse
Affiliation(s)
- M G Dinesh
- Department of Computer Science and Engineering, EASA College of Engineering and Technology, Coimbatore, India
| | | | - S S Askar
- Department of Statistics and Operations Research, College of Science, King Saud University, P.O. Box 2455, 11451, Riyadh, Saudi Arabia
| | - Mohamed Abouhawwash
- Department of Computational Mathematics, Science and Engineering (CMSE), College of Engineering, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Mathematics, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
10
|
Jiang J, Chao WL, Culp S, Krishna SG. Artificial Intelligence in the Diagnosis and Treatment of Pancreatic Cystic Lesions and Adenocarcinoma. Cancers (Basel) 2023; 15:2410. [PMID: 37173876 PMCID: PMC10177524 DOI: 10.3390/cancers15092410] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Pancreatic cancer is projected to become the second leading cause of cancer-related mortality in the United States by 2030. This is in part due to the paucity of reliable screening and diagnostic options for early detection. Amongst known pre-malignant pancreatic lesions, pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasms (IPMNs) are the most prevalent. The current standard of care for the diagnosis and classification of pancreatic cystic lesions (PCLs) involves cross-sectional imaging studies and endoscopic ultrasound (EUS) and, when indicated, EUS-guided fine needle aspiration and cyst fluid analysis. However, this is suboptimal for the identification and risk stratification of PCLs, with accuracy of only 65-75% for detecting mucinous PCLs. Artificial intelligence (AI) is a promising tool that has been applied to improve accuracy in screening for solid tumors, including breast, lung, cervical, and colon cancer. More recently, it has shown promise in diagnosing pancreatic cancer by identifying high-risk populations, risk-stratifying premalignant lesions, and predicting the progression of IPMNs to adenocarcinoma. This review summarizes the available literature on artificial intelligence in the screening and prognostication of precancerous lesions in the pancreas, and streamlining the diagnosis of pancreatic cancer.
Collapse
Affiliation(s)
- Joanna Jiang
- Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Wei-Lun Chao
- Department of Computer Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Stacey Culp
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Somashekar G. Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Ohio State University Wexner Medical Ceter, Columbus, OH 43210, USA
| |
Collapse
|
11
|
Berbís MA, Paulano Godino F, Royuela del Val J, Alcalá Mata L, Luna A. Clinical impact of artificial intelligence-based solutions on imaging of the pancreas and liver. World J Gastroenterol 2023; 29:1427-1445. [PMID: 36998424 PMCID: PMC10044858 DOI: 10.3748/wjg.v29.i9.1427] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/13/2023] [Accepted: 02/27/2023] [Indexed: 03/07/2023] Open
Abstract
Artificial intelligence (AI) has experienced substantial progress over the last ten years in many fields of application, including healthcare. In hepatology and pancreatology, major attention to date has been paid to its application to the assisted or even automated interpretation of radiological images, where AI can generate accurate and reproducible imaging diagnosis, reducing the physicians’ workload. AI can provide automatic or semi-automatic segmentation and registration of the liver and pancreatic glands and lesions. Furthermore, using radiomics, AI can introduce new quantitative information which is not visible to the human eye to radiological reports. AI has been applied in the detection and characterization of focal lesions and diffuse diseases of the liver and pancreas, such as neoplasms, chronic hepatic disease, or acute or chronic pancreatitis, among others. These solutions have been applied to different imaging techniques commonly used to diagnose liver and pancreatic diseases, such as ultrasound, endoscopic ultrasonography, computerized tomography (CT), magnetic resonance imaging, and positron emission tomography/CT. However, AI is also applied in this context to many other relevant steps involved in a comprehensive clinical scenario to manage a gastroenterological patient. AI can also be applied to choose the most convenient test prescription, to improve image quality or accelerate its acquisition, and to predict patient prognosis and treatment response. In this review, we summarize the current evidence on the application of AI to hepatic and pancreatic radiology, not only in regard to the interpretation of images, but also to all the steps involved in the radiological workflow in a broader sense. Lastly, we discuss the challenges and future directions of the clinical application of AI methods.
Collapse
Affiliation(s)
- M Alvaro Berbís
- Department of Radiology, HT Médica, San Juan de Dios Hospital, Córdoba 14960, Spain
- Faculty of Medicine, Autonomous University of Madrid, Madrid 28049, Spain
| | | | | | - Lidia Alcalá Mata
- Department of Radiology, HT Médica, Clínica las Nieves, Jaén 23007, Spain
| | - Antonio Luna
- Department of Radiology, HT Médica, Clínica las Nieves, Jaén 23007, Spain
| |
Collapse
|
12
|
Shao C, Zhang J, Guo J, Zhang L, Zhang Y, Ma L, Gong C, Tian Y, Chen J, Yu N. A radiomics nomogram model for predicting prognosis of pancreatic ductal adenocarcinoma after high-intensity focused ultrasound surgery. Int J Hyperthermia 2023; 40:2184397. [PMID: 36888994 DOI: 10.1080/02656736.2023.2184397] [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: 03/10/2023] Open
Abstract
OBJECTIVE To develop and validate a radiomics nomogram for predicting the survival of patients with pancreatic ductal adenocarcinoma (PDAC) after receiving high-intensity focused ultrasound (HIFU) treatment. METHODS A total of 52 patients with PDAC were enrolled. To select features, the least absolute shrinkage and selection operator algorithm were applied, and the radiomics score (Rad-Score) was obtained. Radiomics model, clinics model, and radiomics nomogram model were constructed by multivariate regression analysis. The identification, calibration, and clinical application of nomogram were evaluated. Survival analysis was performed using Kaplan-Meier (K-M) method. RESULTS According to conclusions made from the multivariate Cox model, Rad-Score, and tumor size were independent risk factors for OS. Compared with the clinical model and radiomics model, the combination of Rad-Score and clinicopathological factors could better predict the survival of patients. Patients were divided into high-risk and low-risk groups according to Rad-Score. K-M analysis showed that the difference between the two groups was statistically significant (p < 0.05). In addition, the radiomics nomogram model indicated better discrimination, calibration, and clinical practicability in training and validation cohorts. CONCLUSION The radiomics nomogram effectively evaluates the prognosis of patients with advanced pancreatic cancer after HIFU surgery, which could potentially improve treatment strategies and promote individualized treatment of advanced pancreatic cancer.
Collapse
Affiliation(s)
- Changjie Shao
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Jing Guo
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liang Zhang
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuhan Zhang
- University of Southern California, Los Angeles, CA, USA
| | - Leiyuan Ma
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chuanxin Gong
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaqi Tian
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingjing Chen
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ning Yu
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
13
|
Hameed BS, Krishnan UM. Artificial Intelligence-Driven Diagnosis of Pancreatic Cancer. Cancers (Basel) 2022; 14:5382. [PMID: 36358800 PMCID: PMC9657087 DOI: 10.3390/cancers14215382] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 08/01/2023] Open
Abstract
Pancreatic cancer is among the most challenging forms of cancer to treat, owing to its late diagnosis and aggressive nature that reduces the survival rate drastically. Pancreatic cancer diagnosis has been primarily based on imaging, but the current state-of-the-art imaging provides a poor prognosis, thus limiting clinicians' treatment options. The advancement of a cancer diagnosis has been enhanced through the integration of artificial intelligence and imaging modalities to make better clinical decisions. In this review, we examine how AI models can improve the diagnosis of pancreatic cancer using different imaging modalities along with a discussion on the emerging trends in an AI-driven diagnosis, based on cytopathology and serological markers. Ethical concerns regarding the use of these tools have also been discussed.
Collapse
Affiliation(s)
- Bahrudeen Shahul Hameed
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), Shanmugha Arts, Science, Technology and Research Academy, Deemed University, Thanjavur 613401, India
- School of Chemical & Biotechnology (SCBT), Shanmugha Arts, Science, Technology and Research Academy, Deemed University, Thanjavur 613401, India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), Shanmugha Arts, Science, Technology and Research Academy, Deemed University, Thanjavur 613401, India
- School of Chemical & Biotechnology (SCBT), Shanmugha Arts, Science, Technology and Research Academy, Deemed University, Thanjavur 613401, India
- School of Arts, Sciences, Humanities & Education (SASHE), Shanmugha Arts, Science, Technology and Research Academy, Deemed University, Thanjavur 613401, India
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Effect of Gray Value Discretization and Image Filtration on Texture Features of the Pancreas Derived from Magnetic Resonance Imaging at 3T. J Imaging 2022; 8:jimaging8080220. [PMID: 36005463 PMCID: PMC9409719 DOI: 10.3390/jimaging8080220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Radiomics of pancreas magnetic resonance (MR) images is positioned well to play an important role in the management of diseases characterized by diffuse involvement of the pancreas. The effect of image pre-processing configurations on these images has been sparsely investigated. Fifteen individuals with definite chronic pancreatitis (an exemplar diffuse disease of the pancreas) and 15 healthy individuals were included in this age- and sex-matched case-control study. MR images of the pancreas were acquired using a single 3T scanner. A total of 93 first-order and second-order texture features of the pancreas were compared between the study groups, by subjecting MR images of the pancreas to 7 image pre-processing configurations related to gray level discretization and image filtration. The studied parameters of intensity discretization did not vary in terms of their effect on the number of significant first-order texture features. The number of statistically significant first-order texture features varied after filtering (7 with the use of logarithm filter and 3 with the use of Laplacian of Gaussian filter with 5 mm σ). Intensity discretization generally affected the number of significant second-order texture features more markedly than filtering. The use of fixed bin number of 16 yielded 42 significant second-order texture features, fixed bin number of 128–38 features, fixed bin width of 6–24 features, and fixed bin width of 42–26 features. The specific parameters of filtration and intensity discretization had differing effects on radiomics signature of the pancreas. Relative discretization with fixed bin number of 16 and use of logarithm filter hold promise as pre-processing configurations of choice in future radiomics studies in diffuse diseases of the pancreas.
Collapse
|
17
|
Laino ME, Ammirabile A, Lofino L, Mannelli L, Fiz F, Francone M, Chiti A, Saba L, Orlandi MA, Savevski V. Artificial Intelligence Applied to Pancreatic Imaging: A Narrative Review. Healthcare (Basel) 2022; 10:healthcare10081511. [PMID: 36011168 PMCID: PMC9408381 DOI: 10.3390/healthcare10081511] [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: 06/20/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 12/19/2022] Open
Abstract
The diagnosis, evaluation, and treatment planning of pancreatic pathologies usually require the combined use of different imaging modalities, mainly, computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET). Artificial intelligence (AI) has the potential to transform the clinical practice of medical imaging and has been applied to various radiological techniques for different purposes, such as segmentation, lesion detection, characterization, risk stratification, or prediction of response to treatments. The aim of the present narrative review is to assess the available literature on the role of AI applied to pancreatic imaging. Up to now, the use of computer-aided diagnosis (CAD) and radiomics in pancreatic imaging has proven to be useful for both non-oncological and oncological purposes and represents a promising tool for personalized approaches to patients. Although great developments have occurred in recent years, it is important to address the obstacles that still need to be overcome before these technologies can be implemented into our clinical routine, mainly considering the heterogeneity among studies.
Collapse
Affiliation(s)
- Maria Elena Laino
- Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
- Correspondence: (M.E.L.); (A.A.)
| | - Angela Ammirabile
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- Department of Diagnostic and Interventional Radiology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
- Correspondence: (M.E.L.); (A.A.)
| | - Ludovica Lofino
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- Department of Diagnostic and Interventional Radiology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | | | - Francesco Fiz
- Nuclear Medicine Unit, Department of Diagnostic Imaging, E.O. Ospedali Galliera, 56321 Genoa, Italy
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital, 72074 Tübingen, Germany
| | - Marco Francone
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- Department of Diagnostic and Interventional Radiology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Arturo Chiti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- Department of Nuclear Medicine, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Luca Saba
- Department of Radiology, University of Cagliari, 09124 Cagliari, Italy
| | | | - Victor Savevski
- Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Schuurmans M, Alves N, Vendittelli P, Huisman H, Hermans J. Setting the Research Agenda for Clinical Artificial Intelligence in Pancreatic Adenocarcinoma Imaging. Cancers (Basel) 2022; 14:cancers14143498. [PMID: 35884559 PMCID: PMC9316850 DOI: 10.3390/cancers14143498] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide, associated with a 98% loss of life expectancy and a 30% increase in disability-adjusted life years. Image-based artificial intelligence (AI) can help improve outcomes for PDAC given that current clinical guidelines are non-uniform and lack evidence-based consensus. However, research on image-based AI for PDAC is too scattered and lacking in sufficient quality to be incorporated into clinical workflows. In this review, an international, multi-disciplinary team of the world’s leading experts in pancreatic cancer breaks down the patient pathway and pinpoints the current clinical touchpoints in each stage. The available PDAC imaging AI literature addressing each pathway stage is then rigorously analyzed, and current performance and pitfalls are identified in a comprehensive overview. Finally, the future research agenda for clinically relevant, image-driven AI in PDAC is proposed. Abstract Pancreatic ductal adenocarcinoma (PDAC), estimated to become the second leading cause of cancer deaths in western societies by 2030, was flagged as a neglected cancer by the European Commission and the United States Congress. Due to lack of investment in research and development, combined with a complex and aggressive tumour biology, PDAC overall survival has not significantly improved the past decades. Cross-sectional imaging and histopathology play a crucial role throughout the patient pathway. However, current clinical guidelines for diagnostic workup, patient stratification, treatment response assessment, and follow-up are non-uniform and lack evidence-based consensus. Artificial Intelligence (AI) can leverage multimodal data to improve patient outcomes, but PDAC AI research is too scattered and lacking in quality to be incorporated into clinical workflows. This review describes the patient pathway and derives touchpoints for image-based AI research in collaboration with a multi-disciplinary, multi-institutional expert panel. The literature exploring AI to address these touchpoints is thoroughly retrieved and analysed to identify the existing trends and knowledge gaps. The results show absence of multi-institutional, well-curated datasets, an essential building block for robust AI applications. Furthermore, most research is unimodal, does not use state-of-the-art AI techniques, and lacks reliable ground truth. Based on this, the future research agenda for clinically relevant, image-driven AI in PDAC is proposed.
Collapse
Affiliation(s)
- Megan Schuurmans
- Diagnostic Image Analysis Group, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; (P.V.); (H.H.)
- Correspondence: (M.S.); (N.A.)
| | - Natália Alves
- Diagnostic Image Analysis Group, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; (P.V.); (H.H.)
- Correspondence: (M.S.); (N.A.)
| | - Pierpaolo Vendittelli
- Diagnostic Image Analysis Group, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; (P.V.); (H.H.)
| | - Henkjan Huisman
- Diagnostic Image Analysis Group, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; (P.V.); (H.H.)
| | - John Hermans
- Department of Medical Imaging, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands;
| |
Collapse
|
21
|
Preuss K, Thach N, Liang X, Baine M, Chen J, Zhang C, Du H, Yu H, Lin C, Hollingsworth MA, Zheng D. Using Quantitative Imaging for Personalized Medicine in Pancreatic Cancer: A Review of Radiomics and Deep Learning Applications. Cancers (Basel) 2022; 14:cancers14071654. [PMID: 35406426 PMCID: PMC8997008 DOI: 10.3390/cancers14071654] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary With a five-year survival rate of only 3% for the majority of patients, pancreatic cancer is a global healthcare challenge. Radiomics and deep learning, two novel quantitative imaging methods that treat medical images as minable data instead of just pictures, have shown promise in advancing personalized management of pancreatic cancer through diagnosing precursor diseases, early detection, accurate diagnosis, and treatment personalization. Radiomics and deep learning methods aim to collect hidden information in medical images that is missed by conventional radiology practices through expanding the data search and comparing information across different patients. Both methods have been studied and applied in pancreatic cancer. In this review, we focus on the current progress of these two methods in pancreatic cancer and provide a comprehensive narrative review on the topic. With better regulation, enhanced workflow, and larger prospective patient datasets, radiomics and deep learning methods could show real hope in the battle against pancreatic cancer through personalized precision medicine. Abstract As the most lethal major cancer, pancreatic cancer is a global healthcare challenge. Personalized medicine utilizing cutting-edge multi-omics data holds potential for major breakthroughs in tackling this critical problem. Radiomics and deep learning, two trendy quantitative imaging methods that take advantage of data science and modern medical imaging, have shown increasing promise in advancing the precision management of pancreatic cancer via diagnosing of precursor diseases, early detection, accurate diagnosis, and treatment personalization and optimization. Radiomics employs manually-crafted features, while deep learning applies computer-generated automatic features. These two methods aim to mine hidden information in medical images that is missed by conventional radiology and gain insights by systematically comparing the quantitative image information across different patients in order to characterize unique imaging phenotypes. Both methods have been studied and applied in various pancreatic cancer clinical applications. In this review, we begin with an introduction to the clinical problems and the technology. After providing technical overviews of the two methods, this review focuses on the current progress of clinical applications in precancerous lesion diagnosis, pancreatic cancer detection and diagnosis, prognosis prediction, treatment stratification, and radiogenomics. The limitations of current studies and methods are discussed, along with future directions. With better standardization and optimization of the workflow from image acquisition to analysis and with larger and especially prospective high-quality datasets, radiomics and deep learning methods could show real hope in the battle against pancreatic cancer through big data-based high-precision personalization.
Collapse
Affiliation(s)
- Kiersten Preuss
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Department of Nutrition and Health Sciences, University of Nebraska Lincoln, Lincoln, NE 68588, USA
| | - Nate Thach
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Department of Computer Science, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Xiaoying Liang
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Michael Baine
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
| | - Justin Chen
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Naperville North High School, Naperville, IL 60563, USA
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Huijing Du
- Department of Mathematics, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Hongfeng Yu
- Department of Computer Science, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Dandan Zheng
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14626, USA
- Correspondence: ; Tel.: +1-(585)-276-3255
| |
Collapse
|
22
|
Janssen BV, Verhoef S, Wesdorp NJ, Huiskens J, de Boer OJ, Marquering H, Stoker J, Kazemier G, Besselink MG. Imaging-based Machine-learning Models to Predict Clinical Outcomes and Identify Biomarkers in Pancreatic Cancer: A Scoping Review. Ann Surg 2022; 275:560-567. [PMID: 34954758 DOI: 10.1097/sla.0000000000005349] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To perform a scoping review of imaging-based machine-learning models to predict clinical outcomes and identify biomarkers in patients with PDAC. SUMMARY OF BACKGROUND DATA Patients with PDAC could benefit from better selection for systemic and surgical therapy. Imaging-based machine-learning models may improve treatment selection. METHODS A scoping review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses-scoping review guidelines in the PubMed and Embase databases (inception-October 2020). The review protocol was prospectively registered (open science framework registration: m4cyx). Included were studies on imaging-based machine-learning models for predicting clinical outcomes and identifying biomarkers for PDAC. The primary outcome was model performance. An area under the curve (AUC) of ≥0.75, or a P-value of ≤0.05, was considered adequate model performance. Methodological study quality was assessed using the modified radiomics quality score. RESULTS After screening 1619 studies, 25 studies with 2305 patients fulfilled the eligibility criteria. All but 1 study was published in 2019 and 2020. Overall, 23/25 studies created models using radiomics features, 1 study quantified vascular invasion on computed tomography, and one used histopathological data. Nine models predicted clinical outcomes with AUC measures of 0.78-0.95, and C-indices of 0.65-0.76. Seventeen models identified biomarkers with AUC measures of 0.68-0.95. Adequate model performance was reported in 23/25 studies. The methodological quality of the included studies was suboptimal, with a median modified radiomics quality score score of 7/36. CONCLUSIONS The use of imaging-based machine-learning models to predict clinical outcomes and identify biomarkers in patients with PDAC is increasingly rapidly. Although these models mostly have good performance scores, their methodological quality should be improved.
Collapse
Affiliation(s)
- Boris V Janssen
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Severano Verhoef
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Nina J Wesdorp
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Onno J de Boer
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Henk Marquering
- Department of Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jaap Stoker
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Geert Kazemier
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Marc G Besselink
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
23
|
High-Resolution, High b-Value Computed Diffusion-Weighted Imaging Improves Detection of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14030470. [PMID: 35158737 PMCID: PMC8833466 DOI: 10.3390/cancers14030470] [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] [Received: 12/28/2021] [Accepted: 01/13/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Our purpose was to investigate the potential of high-resolution, high b-value computed DWI (cDWI) in pancreatic ductal adenocarcinoma (PDAC) detection. Materials and Methods: We retrospectively enrolled 44 patients with confirmed PDAC. Respiratory-triggered, diffusion-weighted, single-shot echo-planar imaging (ss-EPI) with both conventional (i.e., full field-of-view, 3 × 3 × 4 mm voxel size, b = 0, 50, 300, 600 s/mm2) and high-resolution (i.e., reduced field-of-view, 2.5 × 2.5 × 3 mm voxel size, b = 0, 50, 300, 600, 1000 s/mm2) imaging was performed for suspected PDAC. cDWI datasets at b = 1000 s/mm2 were generated for the conventional and high-resolution datasets. Three radiologists were asked to subjectively rate (on a Likert scale of 1–4) the following metrics: image quality, lesion detection and delineation, and lesion-to-pancreas intensity relation. Furthermore, the following quantitative image parameters were assessed: apparent signal-to-noise ratio (aSNR), contrast-to-noise ratio (aCNR), and lesion-to-pancreas contrast ratio (CR). Results: High-resolution, high b-value computed DWI (r-cDWI1000) enabled significant improvement in lesion detection and a higher incidence of a high lesion-to-pancreas intensity relation (type 1, clear hyperintense) compared to conventional high b-value computed and high-resolution high b-value acquired DWI (f-cDWI1000 and r-aDWI1000, respectively). Image quality was rated inferior in the r-cDWI1000 datasets compared to r-aDWI1000. Furthermore, the aCNR and CR were higher in the r-cDWI1000 datasets than in f-cDWI1000 and r-aDWI1000. Conclusion: High-resolution, high b-value computed DWI provides significantly better visualization of PDAC compared to the conventional high b-value computed and high-resolution high b-value images acquired by DWI.
Collapse
|
24
|
Artificial Intelligence in Medicine and Privacy Preservation. Artif Intell Med 2022. [DOI: 10.1007/978-3-030-64573-1_261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
25
|
Casà C, Piras A, D’Aviero A, Preziosi F, Mariani S, Cusumano D, Romano A, Boskoski I, Lenkowicz J, Dinapoli N, Cellini F, Gambacorta MA, Valentini V, Mattiucci GC, Boldrini L. The impact of radiomics in diagnosis and staging of pancreatic cancer. Ther Adv Gastrointest Endosc 2022; 15:26317745221081596. [PMID: 35342883 PMCID: PMC8943316 DOI: 10.1177/26317745221081596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 02/02/2022] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Pancreatic cancer (PC) is one of the most aggressive tumours, and better risk stratification among patients is required to provide tailored treatment. The meaning of radiomics and texture analysis as predictive techniques are not already systematically assessed. The aim of this study is to assess the role of radiomics in PC. METHODS A PubMed/MEDLINE and Embase systematic review was conducted to assess the role of radiomics in PC. The search strategy was 'radiomics [All Fields] AND ("pancreas" [MeSH Terms] OR "pancreas" [All Fields] OR "pancreatic" [All Fields])' and only original articles referred to PC in humans in the English language were considered. RESULTS A total of 123 studies and 183 studies were obtained using the mentioned search strategy on PubMed and Embase, respectively. After the complete selection process, a total of 56 papers were considered eligible for the analysis of the results. Radiomics methods were applied in PC for assessment technical feasibility and reproducibility aspects analysis, risk stratification, biologic or genomic status prediction and treatment response prediction. DISCUSSION Radiomics seems to be a promising approach to evaluate PC from diagnosis to treatment response prediction. Further and larger studies are required to confirm the role and allowed to include radiomics parameter in a comprehensive decision support system.
Collapse
Affiliation(s)
- Calogero Casà
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | | | - Francesco Preziosi
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Silvia Mariani
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Davide Cusumano
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Angela Romano
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ivo Boskoski
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Rome, Italy
| | - Jacopo Lenkowicz
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Nicola Dinapoli
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesco Cellini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Antonietta Gambacorta
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Vincenzo Valentini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gian Carlo Mattiucci
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Boldrini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
26
|
Chen X, Fu R, Shao Q, Chen Y, Ye Q, Li S, He X, Zhu J. Application of artificial intelligence to pancreatic adenocarcinoma. Front Oncol 2022; 12:960056. [PMID: 35936738 PMCID: PMC9353734 DOI: 10.3389/fonc.2022.960056] [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: 06/02/2022] [Accepted: 06/24/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Pancreatic cancer (PC) is one of the deadliest cancers worldwide although substantial advancement has been made in its comprehensive treatment. The development of artificial intelligence (AI) technology has allowed its clinical applications to expand remarkably in recent years. Diverse methods and algorithms are employed by AI to extrapolate new data from clinical records to aid in the treatment of PC. In this review, we will summarize AI's use in several aspects of PC diagnosis and therapy, as well as its limits and potential future research avenues. METHODS We examine the most recent research on the use of AI in PC. The articles are categorized and examined according to the medical task of their algorithm. Two search engines, PubMed and Google Scholar, were used to screen the articles. RESULTS Overall, 66 papers published in 2001 and after were selected. Of the four medical tasks (risk assessment, diagnosis, treatment, and prognosis prediction), diagnosis was the most frequently researched, and retrospective single-center studies were the most prevalent. We found that the different medical tasks and algorithms included in the reviewed studies caused the performance of their models to vary greatly. Deep learning algorithms, on the other hand, produced excellent results in all of the subdivisions studied. CONCLUSIONS AI is a promising tool for helping PC patients and may contribute to improved patient outcomes. The integration of humans and AI in clinical medicine is still in its infancy and requires the in-depth cooperation of multidisciplinary personnel.
Collapse
Affiliation(s)
- Xi Chen
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Ruibiao Fu
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Shao
- Department of Surgical Ward 1, Ningbo Women and Children’s Hospital, Ningbo, China
| | - Yan Chen
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Qinghuang Ye
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng Li
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Xiongxiong He
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Jinhui Zhu
- Department of General Surgery, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Jinhui Zhu,
| |
Collapse
|
27
|
Hayashi H, Uemura N, Matsumura K, Zhao L, Sato H, Shiraishi Y, Yamashita YI, Baba H. Recent advances in artificial intelligence for pancreatic ductal adenocarcinoma. World J Gastroenterol 2021; 27:7480-7496. [PMID: 34887644 PMCID: PMC8613738 DOI: 10.3748/wjg.v27.i43.7480] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 08/02/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains the most lethal type of cancer. The 5-year survival rate for patients with early-stage diagnosis can be as high as 20%, suggesting that early diagnosis plays a pivotal role in the prognostic improvement of PDAC cases. In the medical field, the broad availability of biomedical data has led to the advent of the "big data" era. To overcome this deadly disease, how to fully exploit big data is a new challenge in the era of precision medicine. Artificial intelligence (AI) is the ability of a machine to learn and display intelligence to solve problems. AI can help to transform big data into clinically actionable insights more efficiently, reduce inevitable errors to improve diagnostic accuracy, and make real-time predictions. AI-based omics analyses will become the next alterative approach to overcome this poor-prognostic disease by discovering biomarkers for early detection, providing molecular/genomic subtyping, offering treatment guidance, and predicting recurrence and survival. Advances in AI may therefore improve PDAC survival outcomes in the near future. The present review mainly focuses on recent advances of AI in PDAC for clinicians. We believe that breakthroughs will soon emerge to fight this deadly disease using AI-navigated precision medicine.
Collapse
Affiliation(s)
- Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Norio Uemura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Kazuki Matsumura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Liu Zhao
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Hiroki Sato
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yuta Shiraishi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yo-ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| |
Collapse
|
28
|
Kamal O, McTavish S, Harder FN, Van AT, Peeters JM, Weiss K, Makowski MR, Karampinos DC, Braren RF. Noise reduction in diffusion weighted MRI of the pancreas using an L1-regularized iterative SENSE reconstruction. Magn Reson Imaging 2021; 87:1-6. [PMID: 34808306 DOI: 10.1016/j.mri.2021.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 11/16/2021] [Indexed: 01/19/2023]
Abstract
OBJECTIVES To prospectively evaluate an L1 regularized iterative SENSE reconstruction (L1-R SENSE) to eliminate band-like artifacts frequently seen with parallel imaging (SENSE) at high acceleration factors in high resolution diffusion weighted magnetic resonance imaging of the pancreas. METHODS Fourteen patients with pancreatic ductal adenocarcinoma (PDAC) underwent respiratory triggered DWI ss-EPI at a resolution of 2.5 × 2.5 × 3 mm3 with uniform undersampling in the phase encoding direction (AP axis) with an acceleration factor of 4. Data were reconstructed using the standard SENSE reconstruction routine of the vendor and an iterative SENSE reconstruction employing L1 regularization after a wavelet sparsifying transformation (L1-R SENSE). Retrospective reconstruction of the data with a lower number of averages was performed using both reconstruction methods. Two radiologists independently assessed noise artifacts, anatomical details and image quality (IQ) subjectively with a 4-point scale. Apparent diffusion coefficient (ADC) and covariance (CV) of ADC estimated from images reconstructed at a different number of averages for PDAC and the normal pancreas were assessed. RESULTS L1-R SENSE resulted in higher IQ and less noise artifacts than SENSE. Anatomical details were significantly higher for SENSE in one reader. Mean ADC of PDAC and normal pancreas were significantly higher for L1-R SENSE than SENSE. L1-R SENSE revealed lower CV of ADC for normal pancreas compared to SENSE, whereas no difference was noted for PDAC. CONCLUSION Compared with traditional SENSE reconstruction, L1-R SENSE effectively reduces band-like noise and improves the robustness of the ADC estimation from acquisitions using single-shot DW-EPI of the pancreas.
Collapse
Affiliation(s)
- Omar Kamal
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany; Department of Diagnostic Radiology, Oregon Health and Science University, Oregon, USA
| | - Sean McTavish
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Felix N Harder
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Anh T Van
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany
| | | | | | - Marcus R Makowski
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Dimitrios C Karampinos
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Rickmer F Braren
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Munich partner site, Germany.
| |
Collapse
|
29
|
Harder FN, Kamal O, Kaissis GA, Heid I, Lohöfer FK, McTavish S, Van AT, Katemann C, Peeters JM, Karampinos DC, Makowski MR, Braren RF. Qualitative and Quantitative Comparison of Respiratory Triggered Reduced Field-of-View (FOV) Versus Full FOV Diffusion Weighted Imaging (DWI) in Pancreatic Pathologies. Acad Radiol 2021; 28 Suppl 1:S234-S243. [PMID: 33390324 DOI: 10.1016/j.acra.2020.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 02/08/2023]
Abstract
RATIONALE AND OBJECTIVES To investigate the effects of a reduced field-of-view (rFOV) acquisition in diffusion-weighted magnetic resonance imaging of the pancreas. MATERIALS AND METHODS We enrolled 153 patients who underwent routine clinical MRI work-up including respiratory-triggered diffusion-weighted single-shot echo-planar imaging (DWI) with full field-of-view (fFOV, 3 × 3 × 4 mm3 voxel size) and reduced field-of-view (rFOV, 2.5 × 2.5 × 3 mm3 voxel size) for suspected pancreatic pathology. Two experienced radiologists were asked to subjectively rate (Likert Scale 1-4) image quality (overall image quality, lesion conspicuity, anatomical detail, artifacts). In addition, quantitative image parameters were assessed (apparent diffusion coefficient, apparent signal to noise ratio, apparent contrast to noise ratio [CNR]). RESULTS All subjective metrics of image quality were rated in favor of rFOV DWI images compared to fFOV DWI images with substantial-to-high inter-rater reliability. Calculated ADC values of normal pancreas, pancreatic pathologies and reference tissues revealed no differences between both sequences. Whereas the apparent signal to noise ratio was higher in fFOV images, apparent CNR was higher in rFOV images. CONCLUSION rFOV DWI provides higher image quality and apparent CNR values, favorable in the analysis of pancreatic pathologies.
Collapse
Affiliation(s)
- Felix N Harder
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, School of Medicine
| | - Omar Kamal
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, School of Medicine; Department of Radiology, South Egypt Cancer Institute, Assiut University, Egypt
| | - Georgios A Kaissis
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, School of Medicine; Department of Computing, Faculty of Engineering, Imperial College of Science, Technology and Medicine, United Kingdom
| | - Irina Heid
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, School of Medicine
| | - Fabian K Lohöfer
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, School of Medicine
| | - Sean McTavish
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, School of Medicine
| | - Anh T Van
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, School of Medicine
| | | | | | - Dimitrios C Karampinos
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, School of Medicine
| | - Marcus R Makowski
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, School of Medicine
| | - Rickmer F Braren
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, School of Medicine.
| |
Collapse
|
30
|
Kröner PT, Engels MML, Glicksberg BS, Johnson KW, Mzaik O, van Hooft JE, Wallace MB, El-Serag HB, Krittanawong C. Artificial intelligence in gastroenterology: A state-of-the-art review. World J Gastroenterol 2021; 27:6794-6824. [PMID: 34790008 PMCID: PMC8567482 DOI: 10.3748/wjg.v27.i40.6794] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/15/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
The development of artificial intelligence (AI) has increased dramatically in the last 20 years, with clinical applications progressively being explored for most of the medical specialties. The field of gastroenterology and hepatology, substantially reliant on vast amounts of imaging studies, is not an exception. The clinical applications of AI systems in this field include the identification of premalignant or malignant lesions (e.g., identification of dysplasia or esophageal adenocarcinoma in Barrett’s esophagus, pancreatic malignancies), detection of lesions (e.g., polyp identification and classification, small-bowel bleeding lesion on capsule endoscopy, pancreatic cystic lesions), development of objective scoring systems for risk stratification, predicting disease prognosis or treatment response [e.g., determining survival in patients post-resection of hepatocellular carcinoma), determining which patients with inflammatory bowel disease (IBD) will benefit from biologic therapy], or evaluation of metrics such as bowel preparation score or quality of endoscopic examination. The objective of this comprehensive review is to analyze the available AI-related studies pertaining to the entirety of the gastrointestinal tract, including the upper, middle and lower tracts; IBD; the hepatobiliary system; and the pancreas, discussing the findings and clinical applications, as well as outlining the current limitations and future directions in this field.
Collapse
Affiliation(s)
- Paul T Kröner
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Megan ML Engels
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL 32224, United States
- Cancer Center Amsterdam, Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, Amsterdam 1105, The Netherlands
| | - Benjamin S Glicksberg
- The Hasso Plattner Institute for Digital Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Kipp W Johnson
- The Hasso Plattner Institute for Digital Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Obaie Mzaik
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Amsterdam 2300, The Netherlands
| | - Michael B Wallace
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL 32224, United States
- Division of Gastroenterology and Hepatology, Sheikh Shakhbout Medical City, Abu Dhabi 11001, United Arab Emirates
| | - Hashem B El-Serag
- Section of Gastroenterology and Hepatology, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, TX 77030, United States
- Section of Health Services Research, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, TX 77030, United States
| | - Chayakrit Krittanawong
- Section of Health Services Research, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, TX 77030, United States
- Section of Cardiology, Michael E. DeBakey VA Medical Center, Houston, TX 77030, United States
| |
Collapse
|
31
|
Machine Learning: Applications and Advanced Progresses of Radiomics in Endocrine Neoplasms. JOURNAL OF ONCOLOGY 2021; 2021:8615450. [PMID: 34671399 PMCID: PMC8523238 DOI: 10.1155/2021/8615450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/13/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022]
Abstract
Endocrine neoplasms remain a great threat to human health. It is extremely important to make a clear diagnosis and timely treatment of endocrine tumors. Machine learning includes radiomics, which has long been utilized in clinical cancer research. Radiomics refers to the extraction of valuable information by analyzing a large amount of standard data with high-throughput medical images mainly including computed tomography, positron emission tomography, magnetic resonance imaging, and ultrasound. With the quantitative imaging analysis and model building, radiomics can reflect specific underlying characteristics of a disease that otherwise could not be evaluated visually. More and more promising results of radiomics in oncological practice have been seen in recent years. Radiomics may have the potential to supplement traditional imaging analysis and assist in providing precision medicine for patients. Radiomics had developed rapidly in endocrine neoplasms practice in the past decade. In this review, we would introduce the general workflow of radiomics and summarize the applications and developments of radiomics in endocrine neoplasms in recent years. The limitations of current radiomic research studies and future development directions would also be discussed.
Collapse
|
32
|
Suman G, Patra A, Korfiatis P, Majumder S, Chari ST, Truty MJ, Fletcher JG, Goenka AH. Quality gaps in public pancreas imaging datasets: Implications & challenges for AI applications. Pancreatology 2021; 21:1001-1008. [PMID: 33840636 DOI: 10.1016/j.pan.2021.03.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/15/2021] [Accepted: 03/20/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Quality gaps in medical imaging datasets lead to profound errors in experiments. Our objective was to characterize such quality gaps in public pancreas imaging datasets (PPIDs), to evaluate their impact on previously published studies, and to provide post-hoc labels and segmentations as a value-add for these PPIDs. METHODS We scored the available PPIDs on the medical imaging data readiness (MIDaR) scale, and evaluated for associated metadata, image quality, acquisition phase, etiology of pancreas lesion, sources of confounders, and biases. Studies utilizing these PPIDs were evaluated for awareness of and any impact of quality gaps on their results. Volumetric pancreatic adenocarcinoma (PDA) segmentations were performed for non-annotated CTs by a junior radiologist (R1) and reviewed by a senior radiologist (R3). RESULTS We found three PPIDs with 560 CTs and six MRIs. NIH dataset of normal pancreas CTs (PCT) (n = 80 CTs) had optimal image quality and met MIDaR A criteria but parts of pancreas have been excluded in the provided segmentations. TCIA-PDA (n = 60 CTs; 6 MRIs) and MSD(n = 420 CTs) datasets categorized to MIDaR B due to incomplete annotations, limited metadata, and insufficient documentation. Substantial proportion of CTs from TCIA-PDA and MSD datasets were found unsuitable for AI due to biliary stents [TCIA-PDA:10 (17%); MSD:112 (27%)] or other factors (non-portal venous phase, suboptimal image quality, non-PDA etiology, or post-treatment status) [TCIA-PDA:5 (8.5%); MSD:156 (37.1%)]. These quality gaps were not accounted for in any of the 25 studies that have used these PPIDs (NIH-PCT:20; MSD:1; both: 4). PDA segmentations were done by R1 in 91 eligible CTs (TCIA-PDA:42; MSD:49). Of these, corrections were made by R3 in 16 CTs (18%) (TCIA-PDA:4; MSD:12) [mean (standard deviation) Dice: 0.72(0.21) and 0.63(0.23) respectively]. CONCLUSION Substantial quality gaps, sources of bias, and high proportion of CTs unsuitable for AI characterize the available limited PPIDs. Published studies on these PPIDs do not account for these quality gaps. We complement these PPIDs through post-hoc labels and segmentations for public release on the TCIA portal. Collaborative efforts leading to large, well-curated PPIDs supported by adequate documentation are critically needed to translate the promise of AI to clinical practice.
Collapse
Affiliation(s)
- Garima Suman
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Anurima Patra
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Panagiotis Korfiatis
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Shounak Majumder
- Department of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Suresh T Chari
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Mark J Truty
- Department of General and Gastrointestinal Surgery, Mayo Clinic, 200 First Street SW, MN, 55905, USA
| | - Joel G Fletcher
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
33
|
Chaddad A, Sargos P, Desrosiers C. Modeling Texture in Deep 3D CNN for Survival Analysis. IEEE J Biomed Health Inform 2021; 25:2454-2462. [PMID: 32960772 DOI: 10.1109/jbhi.2020.3025901] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Radiomics has shown remarkable potential for predicting the survival outcome for various types of cancers such as pancreatic ductal adenocarcinoma (PDAC). However, to date, there has been limited research using convolutional neural networks (CNN) with radiomic methods for this task, due to their requirement for large training sets. To overcome this issue, we propose a new type of radiomic descriptor modeling the distribution of learned features with a Gaussian mixture model (GMM). These parametric features (GMM-CNN) are computed from gross tumor volumes of PDAC patients defined semi-automatically in pre-operative computed tomography (CT) scans. We use the proposed GMM-CNN features as input to a robust classifier based on random forests (RF) to predict the survival outcome of patients with PDAC. Our experiments assess the advantage of GMM-CNN compared to employing the same 3D CNN model directly, standard radiomic (i.e., histogram, texture and shape), conditional entropy (CENT) based on 3DCNN, and clinical features (i.e., serum carbohydrate antigen 19-9 and chemotherapy neoadjuvant). Using the RF model (100 samples for training; 59 samples for validation), GMM-CNN features provided the highest area under the ROC curve (AUC) of 72.0% (p = 6.4×10-5) compared to 64.0% (p = 0.01) for the 3D CNN model output, 66.8% (p = 0.01) for standard radiomic features, 64.2% (p = 0.003) for CENT, and 57.6% (p = 0.3) for clinical variables. Our results suggest that the proposed GMM-CNN features used with a RF classifier can significantly improve the capacity to prognosticate PDAC patients prior to surgery via routinely-acquired imaging data.
Collapse
|
34
|
Li X, Yang L, Yuan Z, Lou J, Fan Y, Shi A, Huang J, Zhao M, Wu Y. Multi-institutional development and external validation of machine learning-based models to predict relapse risk of pancreatic ductal adenocarcinoma after radical resection. J Transl Med 2021; 19:281. [PMID: 34193166 PMCID: PMC8243478 DOI: 10.1186/s12967-021-02955-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/19/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Surgical resection is the only potentially curative treatment for pancreatic ductal adenocarcinoma (PDAC) and the survival of patients after radical resection is closely related to relapse. We aimed to develop models to predict the risk of relapse using machine learning methods based on multiple clinical parameters. METHODS Data were collected and analysed of 262 PDAC patients who underwent radical resection at 3 institutions between 2013 and 2017, with 183 from one institution as a training set, 79 from the other 2 institution as a validation set. We developed and compared several predictive models to predict 1- and 2-year relapse risk using machine learning approaches. RESULTS Machine learning techniques were superior to conventional regression-based analyses in predicting risk of relapse of PDAC after radical resection. Among them, the random forest (RF) outperformed other methods in the training set. The highest accuracy and area under the receiver operating characteristic curve (AUROC) for predicting 1-year relapse risk with RF were 78.4% and 0.834, respectively, and for 2-year relapse risk were 95.1% and 0.998. However, the support vector machine (SVM) model showed better performance than the others for predicting 1-year relapse risk in the validation set. And the k neighbor algorithm (KNN) model achieved the highest accuracy and AUROC for predicting 2-year relapse risk. CONCLUSIONS By machine learning, this study has developed and validated comprehensive models integrating clinicopathological characteristics to predict the relapse risk of PDAC after radical resection which will guide the development of personalized surveillance programs after surgery.
Collapse
Affiliation(s)
- Xiawei Li
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, 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, 310000, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Litao Yang
- Department of Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, 310000, Zhejiang, China
| | - Zheping Yuan
- Hessian Health Technology Co., Ltd, Beijing, 100007, China
| | - Jianyao Lou
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, 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, 310000, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yiqun Fan
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
| | - Aiguang Shi
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, 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, 310000, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Junjie Huang
- Department of Surgery, Changxing People's Hospital, Huzhou, 313100, Zhejiang, China
| | - Mingchen Zhao
- Hessian Health Technology Co., Ltd, Beijing, 100007, China
| | - Yulian Wu
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, 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, 310000, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
35
|
Barat M, Chassagnon G, Dohan A, Gaujoux S, Coriat R, Hoeffel C, Cassinotto C, Soyer P. Artificial intelligence: a critical review of current applications in pancreatic imaging. Jpn J Radiol 2021; 39:514-523. [PMID: 33550513 DOI: 10.1007/s11604-021-01098-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022]
Abstract
The applications of artificial intelligence (AI), including machine learning and deep learning, in the field of pancreatic disease imaging are rapidly expanding. AI can be used for the detection of pancreatic ductal adenocarcinoma and other pancreatic tumors but also for pancreatic lesion characterization. In this review, the basic of radiomics, recent developments and current results of AI in the field of pancreatic tumors are presented. Limitations and future perspectives of AI are discussed.
Collapse
Affiliation(s)
- Maxime Barat
- Department of Radiology, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 27 Rue du Faubourg Saint-Jacques, Paris, France
- Université de Paris, Descartes-Paris 5, 75006, Paris, France
| | - Guillaume Chassagnon
- Department of Radiology, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 27 Rue du Faubourg Saint-Jacques, Paris, France
- Université de Paris, Descartes-Paris 5, 75006, Paris, France
| | - Anthony Dohan
- Department of Radiology, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 27 Rue du Faubourg Saint-Jacques, Paris, France
- Université de Paris, Descartes-Paris 5, 75006, Paris, France
| | - Sébastien Gaujoux
- Université de Paris, Descartes-Paris 5, 75006, Paris, France
- Department of Abdominal Surgery, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 75014, Paris, France
| | - Romain Coriat
- Université de Paris, Descartes-Paris 5, 75006, Paris, France
- Department of Gastroenterology, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 75014, Paris, France
| | - Christine Hoeffel
- Department of Radiology, Robert Debré Hospital, 51092, Reims, France
| | - Christophe Cassinotto
- Department of Radiology, CHU Montpellier, University of Montpellier, Saint-Éloi Hospital, 34000, Montpellier, France
| | - Philippe Soyer
- Department of Radiology, Hopital Cochin, Assistance Publique-Hopitaux de Paris, 27 Rue du Faubourg Saint-Jacques, Paris, France.
- Université de Paris, Descartes-Paris 5, 75006, Paris, France.
| |
Collapse
|
36
|
Traub B, Link KH, Kornmann M. Curing pancreatic cancer. Semin Cancer Biol 2021; 76:232-246. [PMID: 34062264 DOI: 10.1016/j.semcancer.2021.05.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/14/2022]
Abstract
The distinct biology of pancreatic cancer with aggressive and early invasive tumor cells, a tumor promoting microenvironment, late diagnosis, and high therapy resistance poses major challenges on clinicians, researchers, and patients. In current clinical practice, a curative approach for pancreatic cancer can only be offered to a minority of patients and even for those patients, the long-term outcome is grim. This bitter combination will eventually let pancreatic cancer rise to the second leading cause of cancer-related mortalities. With surgery being the only curative option, complete tumor resection still remains the center of pancreatic cancer treatment. In recent years, new developments in neoadjuvant and adjuvant treatment have emerged. Together with improved perioperative care including complication management, an increasing number of patients have become eligible for tumor resection. Basic research aims to further increase these numbers by new methods of early detection, better tumor modelling and personalized treatment options. This review aims to summarize the current knowledge on clinical and biologic features, surgical and non-surgical treatment options, and the improved collaboration of clinicians and basic researchers in pancreatic cancer that will hopefully result in more successful ways of curing pancreatic cancer.
Collapse
Affiliation(s)
- Benno Traub
- Clinic for General and Visceral Surgery, University of Ulm, Albert-Einstein Allee 23, Ulm, Germany.
| | - Karl-Heinz Link
- Clinic for General and Visceral Surgery, University of Ulm, Ulm, Germany; Surgical and Asklepios Tumor Center (ATC), Asklepios Paulinen Klinik Wiesbaden, Richard Strauss-Str. 4, Wiesbaden, Germany.
| | - Marko Kornmann
- Clinic for General and Visceral Surgery, University of Ulm, Albert-Einstein Allee 23, Ulm, Germany.
| |
Collapse
|
37
|
Mendoza Ladd A, Diehl DL. Artificial intelligence for early detection of pancreatic adenocarcinoma: The future is promising. World J Gastroenterol 2021; 27:1283-1295. [PMID: 33833482 PMCID: PMC8015296 DOI: 10.3748/wjg.v27.i13.1283] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/22/2021] [Accepted: 03/13/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a worldwide public health concern. Despite extensive research efforts toward improving diagnosis and treatment, the 5-year survival rate at best is approximately 15%. This dismal figure can be attributed to a variety of factors including lack of adequate screening methods, late symptom onset, and treatment resistance. Pancreatic ductal adenocarcinoma remains a grim diagnosis with a high mortality rate and a significant psy-chological burden for patients and their families. In recent years artificial intelligence (AI) has permeated the medical field at an accelerated pace, bringing potential new tools that carry the promise of improving diagnosis and treatment of a variety of diseases. In this review we will summarize the landscape of AI in diagnosis and treatment of PDAC.
Collapse
Affiliation(s)
- Antonio Mendoza Ladd
- Department of Internal Medicine, Division of Gastroenterology, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, United States
| | - David L Diehl
- Department of Gastroenterology and Nutrition, Geisinger Medical Center, Danville, PA 17822, United States
| |
Collapse
|
38
|
Xing H, Hao Z, Zhu W, Sun D, Ding J, Zhang H, Liu Y, Huo L. Preoperative prediction of pathological grade in pancreatic ductal adenocarcinoma based on 18F-FDG PET/CT radiomics. EJNMMI Res 2021; 11:19. [PMID: 33630176 PMCID: PMC7907291 DOI: 10.1186/s13550-021-00760-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/10/2021] [Indexed: 12/19/2022] Open
Abstract
Purpose To develop and validate a machine learning model based on radiomic features derived from 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) images to preoperatively predict the pathological grade in patients with pancreatic ductal adenocarcinoma (PDAC). Methods A total of 149 patients (83 men, 66 women, mean age 61 years old) with pathologically proven PDAC and a preoperative 18F-FDG PET/CT scan between May 2009 and January 2016 were included in this retrospective study. The cohort of patients was divided into two separate groups for the training (99 patients) and validation (50 patients) in chronological order. Radiomics features were extracted from PET/CT images using Pyradiomics implemented in Python, and the XGBoost algorithm was used to build a prediction model. Conventional PET parameters, including standardized uptake value, metabolic tumor volume, and total lesion glycolysis, were also measured. The quality of the proposed model was appraised by means of receiver operating characteristics (ROC) and areas under the ROC curve (AUC). Results The prediction model based on a twelve-feature-combined radiomics signature could stratify PDAC patients into grade 1 and grade 2/3 groups with AUC of 0.994 in the training set and 0.921 in the validation set. Conclusion The model developed is capable of predicting pathological differentiation grade of PDAC based on preoperative 18F-FDG PET/CT radiomics features.
Collapse
Affiliation(s)
- Haiqun Xing
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Dongcheng District, Chinese Academy of Medical Science, Peking Union Medical College, No.1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Zhixin Hao
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Dongcheng District, Chinese Academy of Medical Science, Peking Union Medical College, No.1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Wenjia Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Dongcheng District, Chinese Academy of Medical Science, Peking Union Medical College, No.1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Dehui Sun
- Sinounion Healthcare Inc., Building 3-B, Zhongguancun Dong Sheng International Pioneer Park, Beijing, 100192, China
| | - Jie Ding
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Dongcheng District, Chinese Academy of Medical Science, Peking Union Medical College, No.1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yu Liu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Dongcheng District, Chinese Academy of Medical Science, Peking Union Medical College, No.1 Shuaifuyuan, Beijing, 100730, China.,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Dongcheng District, Chinese Academy of Medical Science, Peking Union Medical College, No.1 Shuaifuyuan, Beijing, 100730, China. .,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China.
| |
Collapse
|
39
|
Cusumano D, Boldrini L, Yadav P, Casà C, Lee SL, Romano A, Piras A, Chiloiro G, Placidi L, Catucci F, Votta C, Mattiucci GC, Indovina L, Gambacorta MA, Bassetti M, Valentini V. Delta Radiomics Analysis for Local Control Prediction in Pancreatic Cancer Patients Treated Using Magnetic Resonance Guided Radiotherapy. Diagnostics (Basel) 2021; 11:diagnostics11010072. [PMID: 33466307 PMCID: PMC7824764 DOI: 10.3390/diagnostics11010072] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/31/2020] [Accepted: 12/31/2020] [Indexed: 02/07/2023] Open
Abstract
The aim of this study is to investigate the role of Delta Radiomics analysis in the prediction of one-year local control (1yLC) in patients affected by locally advanced pancreatic cancer (LAPC) and treated using Magnetic Resonance guided Radiotherapy (MRgRT). A total of 35 patients from two institutions were enrolled: A 0.35 Tesla T2*/T1 MR image was acquired for each case during simulation and on each treatment fraction. Physical dose was converted in biologically effective dose (BED) to compensate for different radiotherapy schemes. Delta Radiomics analysis was performed considering the gross tumour volume (GTV) delineated on MR images acquired at BED of 20, 40, and 60 Gy. The performance of the delta features in predicting 1yLC was investigated in terms of Wilcoxon Mann-Whitney test and area under receiver operating characteristic (ROC) curve (AUC). The most significant feature in predicting 1yLC was the variation of cluster shade calculated at BED = 40 Gy, with a p-value of 0.005 and an AUC of 0.78 (0.61-0.94). Delta Radiomics analysis on low-field MR images might play a promising role in 1yLC prediction for LAPC patients: further studies including an external validation dataset and a larger cohort of patients are recommended to confirm the validity of this preliminary experience.
Collapse
Affiliation(s)
- Davide Cusumano
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| | - Luca Boldrini
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| | - Poonam Yadav
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave, Madison, WI 53792, USA; (P.Y.); (M.B.)
| | - Calogero Casà
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
- Correspondence: ; Tel.: +39-06-3015-5226
| | - Sangjune Laurence Lee
- Department of Oncology, University of Calgary, 1331 29 Street NW, Calgary, AB T2N 1N4, Canada;
| | - Angela Romano
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| | - Antonio Piras
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| | - Giuditta Chiloiro
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| | - Lorenzo Placidi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| | - Francesco Catucci
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| | - Claudio Votta
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| | - Gian Carlo Mattiucci
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| | - Luca Indovina
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| | - Maria Antonietta Gambacorta
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| | - Michael Bassetti
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave, Madison, WI 53792, USA; (P.Y.); (M.B.)
| | - Vincenzo Valentini
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (D.C.); (L.B.); (A.R.); (A.P.); (G.C.); (L.P.); (F.C.); (C.V.); (G.C.M.); (L.I.); (M.A.G.); (V.V.)
| |
Collapse
|
40
|
Akshintala VS, Khashab MA. Artificial intelligence in pancreaticobiliary endoscopy. J Gastroenterol Hepatol 2021; 36:25-30. [PMID: 33448514 DOI: 10.1111/jgh.15343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023]
Abstract
Artificial intelligence (AI) applications in health care have exponentially increased in recent years, and a few of these are related to pancreatobiliary disorders. AI-based methods were applied to extract information, in prognostication, to guide clinical treatment decisions and in pancreatobiliary endoscopy to characterize lesions. AI applications in endoscopy are expected to reduce inter-operator variability, improve the accuracy of diagnosis, and assist in therapeutic decision-making in real time. AI-based literature must however be interpreted with caution given the limited external validation. A multidisciplinary approach combining clinical and imaging or endoscopy data will better utilize AI-based technologies to further improve patient care.
Collapse
Affiliation(s)
- Venkata S Akshintala
- Division of Gastroenterology, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Mouen A Khashab
- Division of Gastroenterology, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| |
Collapse
|
41
|
Gorris M, Hoogenboom SA, Wallace MB, van Hooft JE. Artificial intelligence for the management of pancreatic diseases. Dig Endosc 2021; 33:231-241. [PMID: 33065754 PMCID: PMC7898901 DOI: 10.1111/den.13875] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/29/2020] [Accepted: 10/11/2020] [Indexed: 12/16/2022]
Abstract
Novel artificial intelligence techniques are emerging in all fields of healthcare, including gastroenterology. The aim of this review is to give an overview of artificial intelligence applications in the management of pancreatic diseases. We performed a systematic literature search in PubMed and Medline up to May 2020 to identify relevant articles. Our results showed that the development of machine-learning based applications is rapidly evolving in the management of pancreatic diseases, guiding precision medicine in clinical, endoscopic and radiologic settings. Before implementation into clinical practice, further research should focus on the external validation of novel techniques, clarifying the accuracy and robustness of these models.
Collapse
Affiliation(s)
- Myrte Gorris
- Department of Gastroenterology and HepatologyAmsterdam Gastroenterology Endocrinology MetabolismAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Sanne A. Hoogenboom
- Department of Gastroenterology and HepatologyAmsterdam Gastroenterology Endocrinology MetabolismAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Michael B. Wallace
- Department of Gastroenterology and HepatologyMayo Clinic JacksonvilleJacksonvilleUSA
| | - Jeanin E. van Hooft
- Department of Gastroenterology and HepatologyAmsterdam Gastroenterology Endocrinology MetabolismAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Gastroenterology and HepatologyLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
42
|
Artificial Intelligence in Medicine and Privacy Preservation. Artif Intell Med 2021. [DOI: 10.1007/978-3-030-58080-3_261-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
43
|
The integration of artificial intelligence models to augment imaging modalities in pancreatic cancer. JOURNAL OF PANCREATOLOGY 2020. [DOI: 10.1097/jp9.0000000000000056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
44
|
Abunahel BM, Pontre B, Kumar H, Petrov MS. Pancreas image mining: a systematic review of radiomics. Eur Radiol 2020; 31:3447-3467. [PMID: 33151391 DOI: 10.1007/s00330-020-07376-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/25/2020] [Accepted: 10/05/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To systematically review published studies on the use of radiomics of the pancreas. METHODS The search was conducted in the MEDLINE database. Human studies that investigated the applications of radiomics in diseases of the pancreas were included. The radiomics quality score was calculated for each included study. RESULTS A total of 72 studies encompassing 8863 participants were included. Of them, 66 investigated focal pancreatic lesions (pancreatic cancer, precancerous lesions, or benign lesions); 4, pancreatitis; and 2, diabetes mellitus. The principal applications of radiomics were differential diagnosis between various types of focal pancreatic lesions (n = 19), classification of pancreatic diseases (n = 23), and prediction of prognosis or treatment response (n = 30). Second-order texture features were most useful for the purpose of differential diagnosis of diseases of the pancreas (with 100% of studies investigating them found a statistically significant feature), whereas filtered image features were most useful for the purpose of classification of diseases of the pancreas and prediction of diseases of the pancreas (with 100% of studies investigating them found a statistically significant feature). The median radiomics quality score of the included studies was 28%, with the interquartile range of 22% to 36%. The radiomics quality score was significantly correlated with the number of extracted radiomics features (r = 0.52, p < 0.001) and the study sample size (r = 0.34, p = 0.003). CONCLUSIONS Radiomics of the pancreas holds promise as a quantitative imaging biomarker of both focal pancreatic lesions and diffuse changes of the pancreas. The usefulness of radiomics features may vary depending on the purpose of their application. Standardisation of image acquisition protocols and image pre-processing is warranted prior to considering the use of radiomics of the pancreas in routine clinical practice. KEY POINTS • Methodologically sound studies on radiomics of the pancreas are characterised by a large sample size and a large number of extracted features. • Optimisation of the radiomics pipeline will increase the clinical utility of mineable pancreas imaging data. • Radiomics of the pancreas is a promising personalised medicine tool in diseases of the pancreas.
Collapse
Affiliation(s)
| | - Beau Pontre
- School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Haribalan Kumar
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Maxim S Petrov
- School of Medicine, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
45
|
van Wijk L, de Klein GW, Kanters MA, Patijn GA, Klaase JM. The ultimate preoperative C-reactive protein-to-albumin ratio is a prognostic factor for survival after pancreatic cancer resection. Eur J Med Res 2020; 25:46. [PMID: 33028394 PMCID: PMC7541315 DOI: 10.1186/s40001-020-00444-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/15/2020] [Indexed: 12/16/2022] Open
Abstract
Background Emerging evidence indicates that an elevated C-reactive protein-to-albumin ratio (CAR) may be associated with a poor prognosis in pancreatic ductal adenocarcinoma (PDAC). Further evidence showing that this ratio has significant prognostic value could contribute to current prediction models and clinical decision-making. Methods Data were analysed of consecutive patients who underwent curative pancreatic resection between 2013 and 2018 and were histologically diagnosed with PDAC. We investigated the relation between the ultimate preoperative CAR and overall survival. Results A total of 163 patients were analysed. Median overall survival was 18 months (IQR 9–36). Multivariate analysis demonstrated that a higher CAR (HR 1.745, P = 0.004), a higher age (HR 1.062, P < 0.001), male sex (HR 1.977, P = 0.001), poor differentiation grade (HR 2.812, P < 0.001), and positive para-aortic lymph node(s) (HR 4.489, P < 0.001) were associated with a lower overall survival. Furthermore, a CAR ≥ 0.2 was associated with decreased overall survival (16 vs. 26 months, P = 0.003). Conclusion We demonstrated that an ultimate preoperative elevated CAR is an independent indicator of decreased overall survival after resection for PDAC. The preoperative CAR may be of additional value to the current prediction models.
Collapse
Affiliation(s)
- Laura van Wijk
- Department of Hepatobiliary Surgery and Liver Transplantation, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, Groningen, 9700 RB, The Netherlands
| | - Guus W de Klein
- Department of Surgery, Isala, PO Box 10400, Dokter van Heesweg 2, Zwolle, 8000 GK, The Netherlands
| | - Matthijs A Kanters
- Department of Hepatobiliary Surgery and Liver Transplantation, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, Groningen, 9700 RB, The Netherlands
| | - Gijs A Patijn
- Department of Surgery, Isala, PO Box 10400, Dokter van Heesweg 2, Zwolle, 8000 GK, The Netherlands
| | - Joost M Klaase
- Department of Hepatobiliary Surgery and Liver Transplantation, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, Groningen, 9700 RB, The Netherlands.
| |
Collapse
|
46
|
Peng Y, Lin P, Wu L, Wan D, Zhao Y, Liang L, Ma X, Qin H, Liu Y, Li X, Wang X, He Y, Yang H. Ultrasound-Based Radiomics Analysis for Preoperatively Predicting Different Histopathological Subtypes of Primary Liver Cancer. Front Oncol 2020; 10:1646. [PMID: 33072550 PMCID: PMC7543652 DOI: 10.3389/fonc.2020.01646] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Background Preoperative identification of hepatocellular carcinoma (HCC), combined hepatocellular–cholangiocarcinoma (cHCC-ICC), and intrahepatic cholangiocarcinoma (ICC) is essential for treatment decision making. We aimed to use ultrasound-based radiomics analysis to non-invasively distinguish histopathological subtypes of primary liver cancer (PLC) before surgery. Methods We retrospectively analyzed ultrasound images of 668 PLC patients, comprising 531 HCC patients, 48 cHCC-ICC patients, and 89 ICC patients. The boundary of a tumor was manually determined on the largest imaging slice of the ultrasound medicine image by ITK-SNAP software (version 3.8.0), and then, the high-throughput radiomics features were extracted from the obtained region of interest (ROI) of the tumor. The combination of different dimension-reduction technologies and machine learning approaches was used to identify important features and develop the moderate radiomics model. The comprehensive ability of the radiomics model can be evaluated by the area under the receiver operating characteristic curve (AUC). Results After digitally processing tumor ultrasound images, 5,234 high-throughput radiomics features were obtained. We used the Spearman + least absolute shrinkage and selection operator (LASSO) regression method for feature selection and logistics regression for modeling to develop the HCC-vs-non-HCC radiomics model (composed of 16 features). The Spearman + statistical test + random forest methods were used for feature selection, and logistics regression was applied for modeling to develop the ICC-vs-cHCC-ICC radiomics model (composed of 19 features). The overall performance of the radiomics model in identifying different histopathological types of PLC was moderate, with AUC values of 0.854 (training cohort) and 0.775 (test cohort) in the HCC-vs-non-HCC radiomics model and 0.920 (training cohort) and 0.728 (test cohort) in the ICC-vs-cHCC-ICC radiomics model. Conclusion Ultrasound-based radiomics models can help distinguish histopathological subtypes of PLC and provide effective clinical decision making for the accurate diagnosis and treatment of PLC.
Collapse
Affiliation(s)
- Yuting Peng
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Peng Lin
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Linyong Wu
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Da Wan
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yujia Zhao
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Liang
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaoyu Ma
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hui Qin
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yichen Liu
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xin Li
- GE Healthcare, Shanghai, China
| | | | - Yun He
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hong Yang
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
47
|
Ren L, Mota Reyes C, Friess H, Demir IE. Neoadjuvant therapy in pancreatic cancer: what is the true oncological benefit? Langenbecks Arch Surg 2020; 405:879-887. [PMID: 32776259 PMCID: PMC7541356 DOI: 10.1007/s00423-020-01946-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Neoadjuvant therapies (neoTx) have revolutionized the treatment of borderline resectable (BR) and locally advanced (LA) pancreatic cancer (PCa) by significantly increasing the rate of R0 resections, which remains the only curative strategy for these patients. However, there is still room for improvement of neoTx in PCa. PURPOSE Here, we aimed to critically analyze the benefits of neoTx in LA and BR PCa and its potential use on patients with resectable PCa. We also explored the feasibility of arterial resection (AR) to increase surgical radicality and the incorporation of immunotherapy to optimize neoadjuvant approaches in PCa. CONCLUSION For early stage, i.e., resectable, PCa, there is not enough scientific evidence for routinely recommending neoTx. For LA and BR PCa, optimization of neoadjuvant therapy necessitates more sophisticated complex surgical resections, machine learning and radiomic approaches, integration of immunotherapy due to the high antigen load, standardized histopathological assessment, and improved multidisciplinary communication.
Collapse
Affiliation(s)
- Lei Ren
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str. 22, D-81675, Munich, Germany
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Carmen Mota Reyes
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str. 22, D-81675, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str. 22, D-81675, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str. 22, D-81675, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
- CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany.
- Department of General Surgery, HPB Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.
| |
Collapse
|
48
|
Kaissis GA, Makowski MR, Rückert D, Braren RF. Secure, privacy-preserving and federated machine learning in medical imaging. NAT MACH INTELL 2020. [DOI: 10.1038/s42256-020-0186-1] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
49
|
Kaissis GA, Jungmann F, Ziegelmayer S, Lohöfer FK, Harder FN, Schlitter AM, Muckenhuber A, Steiger K, Schirren R, Friess H, Schmid R, Weichert W, Makowski MR, Braren RF. Multiparametric Modelling of Survival in Pancreatic Ductal Adenocarcinoma Using Clinical, Histomorphological, Genetic and Image-Derived Parameters. J Clin Med 2020; 9:jcm9051250. [PMID: 32344944 PMCID: PMC7287805 DOI: 10.3390/jcm9051250] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023] Open
Abstract
Rationale: Pancreatic ductal adenocarcinoma (PDAC) remains a tumor entity of exceptionally poor prognosis, and several biomarkers are under current investigation for the prediction of patient prognosis. Many studies focus on promoting newly developed imaging biomarkers without a rigorous comparison to other established parameters. To assess the true value and leverage the potential of all efforts in this field, a multi-parametric evaluation of the available biomarkers for PDAC survival prediction is warranted. Here we present a multiparametric analysis to assess the predictive value of established parameters and the added contribution of newly developed imaging features such as biomarkers for overall PDAC patient survival. Methods: 103 patients with resectable PDAC were retrospectively enrolled. Clinical and histopathological data (age, sex, chemotherapy regimens, tumor size, lymph node status, grading and resection status), morpho-molecular and genetic data (tumor morphology, molecular subtype, tp53, kras, smad4 and p16 genetics), image-derived features and the combination of all parameters were tested for their prognostic strength based on the concordance index (CI) of multivariate Cox proportional hazards survival modelling after unsupervised machine learning preprocessing. Results: The average CIs of the out-of-sample data were: 0.63 for the clinical and histopathological features, 0.53 for the morpho-molecular and genetic features, 0.65 for the imaging features and 0.65 for the combined model including all parameters. Conclusions: Imaging-derived features represent an independent survival predictor in PDAC and enable the multiparametric, machine learning-assisted modelling of postoperative overall survival with a high performance compared to clinical and morpho-molecular/genetic parameters. We propose that future studies systematically include imaging-derived features to benchmark their additive value when evaluating biomarker-based model performance.
Collapse
Affiliation(s)
- Georgios A. Kaissis
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Department of Computing, Faculty of Engineering, Imperial College of Science, Technology and Medicine, London SW7 2BU, UK
| | - Friederike Jungmann
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Sebastian Ziegelmayer
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Fabian K. Lohöfer
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Felix N. Harder
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Anna Melissa Schlitter
- Institute for Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- German Cancer Consortium, Partner Site Technical University of Munich, D-69120 Heidelberg, Germany
| | - Alexander Muckenhuber
- Institute for Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Katja Steiger
- Institute for Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Rebekka Schirren
- School of Medicine, Surgical Clinic and Policlinic, Technical University of Munich, 81675 Munich, Germany
| | - Helmut Friess
- School of Medicine, Surgical Clinic and Policlinic, Technical University of Munich, 81675 Munich, Germany
| | - Roland Schmid
- Department of Internal Medicine II, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Wilko Weichert
- Institute for Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Marcus R. Makowski
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Rickmer F. Braren
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-4140-5627
| |
Collapse
|
50
|
Kaissis GA, Ziegelmayer S, Lohöfer FK, Harder FN, Jungmann F, Sasse D, Muckenhuber A, Yen HY, Steiger K, Siveke J, Friess H, Schmid R, Weichert W, Makowski MR, Braren RF. Image-Based Molecular Phenotyping of Pancreatic Ductal Adenocarcinoma. J Clin Med 2020; 9:jcm9030724. [PMID: 32155990 PMCID: PMC7141256 DOI: 10.3390/jcm9030724] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
To bridge the translational gap between recent discoveries of distinct molecular phenotypes of pancreatic cancer and tangible improvements in patient outcome, there is an urgent need to develop strategies and tools informing and improving the clinical decision process. Radiomics and machine learning approaches can offer non-invasive whole tumor analytics for clinical imaging data-based classification. The retrospective study assessed baseline computed tomography (CT) from 207 patients with proven pancreatic ductal adenocarcinoma (PDAC). Following expert level manual annotation, Pyradiomics was used for the extraction of 1474 radiomic features. The molecular tumor subtype was defined by immunohistochemical staining for KRT81 and HNF1a as quasi-mesenchymal (QM) vs. non-quasi-mesenchymal (non-QM). A Random Forest machine learning algorithm was developed to predict the molecular subtype from the radiomic features. The algorithm was then applied to an independent cohort of histopathologically unclassifiable tumors with distinct clinical outcomes. The classification algorithm achieved a sensitivity, specificity and ROC-AUC (area under the receiver operating characteristic curve) of 0.84 ± 0.05, 0.92 ± 0.01 and 0.93 ± 0.01, respectively. The median overall survival for predicted QM and non-QM tumors was 16.1 and 20.9 months, respectively, log-rank-test p = 0.02, harzard ratio (HR) 1.59. The application of the algorithm to histopathologically unclassifiable tumors revealed two groups with significantly different survival (8.9 and 39.8 months, log-rank-test p < 0.001, HR 4.33). The machine learning-based analysis of preoperative (CT) imaging allows the prediction of molecular PDAC subtypes highly relevant for patient survival, allowing advanced pre-operative patient stratification for precision medicine applications.
Collapse
Affiliation(s)
- Georgios A. Kaissis
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Radiology, 81675 Munich, Germany; (G.A.K.); (S.Z.); (F.K.L.); (F.N.H.); (F.J.); (D.S.); (M.R.M.)
- Imperial College of Science, Technology and Medicine, Faculty of Engineering, Department of Computing, SW7 2AZ London, UK
| | - Sebastian Ziegelmayer
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Radiology, 81675 Munich, Germany; (G.A.K.); (S.Z.); (F.K.L.); (F.N.H.); (F.J.); (D.S.); (M.R.M.)
| | - Fabian K. Lohöfer
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Radiology, 81675 Munich, Germany; (G.A.K.); (S.Z.); (F.K.L.); (F.N.H.); (F.J.); (D.S.); (M.R.M.)
| | - Felix N. Harder
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Radiology, 81675 Munich, Germany; (G.A.K.); (S.Z.); (F.K.L.); (F.N.H.); (F.J.); (D.S.); (M.R.M.)
| | - Friederike Jungmann
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Radiology, 81675 Munich, Germany; (G.A.K.); (S.Z.); (F.K.L.); (F.N.H.); (F.J.); (D.S.); (M.R.M.)
| | - Daniel Sasse
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Radiology, 81675 Munich, Germany; (G.A.K.); (S.Z.); (F.K.L.); (F.N.H.); (F.J.); (D.S.); (M.R.M.)
| | - Alexander Muckenhuber
- Technical University of Munich, School of Medicine, Institute for Pathology, 81675 Munich, Germany; (A.M.); (H.-Y.Y.); (K.S.); (W.W.)
| | - Hsi-Yu Yen
- Technical University of Munich, School of Medicine, Institute for Pathology, 81675 Munich, Germany; (A.M.); (H.-Y.Y.); (K.S.); (W.W.)
| | - Katja Steiger
- Technical University of Munich, School of Medicine, Institute for Pathology, 81675 Munich, Germany; (A.M.); (H.-Y.Y.); (K.S.); (W.W.)
| | - Jens Siveke
- Institute of Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, 45147 Essen, Germany;
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, parter site Essen, Germany) and German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Helmut Friess
- Technical University of Munich, School of Medicine, Surgical Clinic and Policlinic, 81675 Munich, Germany;
| | - Roland Schmid
- Technical University of Munich, School of Medicine, Department of Internal Medicine II, 81675 Munich, Germany;
| | - Wilko Weichert
- Technical University of Munich, School of Medicine, Institute for Pathology, 81675 Munich, Germany; (A.M.); (H.-Y.Y.); (K.S.); (W.W.)
| | - Marcus R. Makowski
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Radiology, 81675 Munich, Germany; (G.A.K.); (S.Z.); (F.K.L.); (F.N.H.); (F.J.); (D.S.); (M.R.M.)
| | - Rickmer F. Braren
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Radiology, 81675 Munich, Germany; (G.A.K.); (S.Z.); (F.K.L.); (F.N.H.); (F.J.); (D.S.); (M.R.M.)
- Correspondence: ; Tel.: +49-89-4140-5627
| |
Collapse
|