1
|
Zou Y, Xie Y, Huang J, Liang Y, Chang S, Wang H, Wang Y, Gao C, Wang X, Zhao T, Yu J, Gao S, Hao J. Survival outcomes of adjuvant chemotherapy in patients with stage I pancreatic cancer stratified by pathologic risk. Surgery 2024; 176:1466-1474. [PMID: 39191600 DOI: 10.1016/j.surg.2024.07.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/16/2024] [Accepted: 07/27/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND This study aimed to evaluate the impact of adjuvant chemotherapy on survival in patients with stage I pancreatic ductal adenocarcinoma, stratified according to pathologic risk factors. METHODS A total of 259 patients with stage I pancreatic ductal adenocarcinoma who underwent radical resection were retrospectively analyzed. The patients were categorized into groups with and without pathologic risk based on the presence of perineural and/or lymphovascular invasion. Subset Kaplan-Meier survival and multivariate analyses were performed to determine the recurrence-free survival and overall survival. RESULTS Adjuvant chemotherapy did not significantly prolong recurrence-free survival (P = .213) but increased overall survival (P = .019) in patients with stage I pancreatic ductal adenocarcinoma. In subgroup analysis, adjuvant chemotherapy significantly improved recurrence-free survival (P = .037) and overall survival (P = .007) in patients with pathologic risk (n = 160). However, patients without pathologic risk (n = 99) showed no enhancement of recurrence-free survival (P = .870) and overall survival (P = .413) after adjuvant chemotherapy. Subset multivariate analyses indicated that adjuvant chemotherapy was an independent favorable prognostic factor in patients with pathologic risk but not in those without pathologic risk. CONCLUSION Adjuvant chemotherapy for stage I pancreatic ductal adenocarcinoma may provide survival benefits specifically to patients with pathologic risk.
Collapse
Affiliation(s)
- Yiping Zou
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yongjie Xie
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jing Huang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yuexiang Liang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Shaofei Chang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hongwei Wang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yifei Wang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chuntao Gao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiuchao Wang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tiansuo Zhao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jun Yu
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Song Gao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jihui Hao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
2
|
Huang C, Shen Y, Galgano SJ, Goenka AH, Hecht EM, Kambadakone A, Wang ZJ, Chu LC. Advancements in early detection of pancreatic cancer: the role of artificial intelligence and novel imaging techniques. Abdom Radiol (NY) 2024:10.1007/s00261-024-04644-7. [PMID: 39467913 DOI: 10.1007/s00261-024-04644-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024]
Abstract
Early detection is crucial for improving survival rates of pancreatic ductal adenocarcinoma (PDA), yet current diagnostic methods can often fail at this stage. Recently, there has been significant interest in improving risk stratification and developing imaging biomarkers, through novel imaging techniques, and most notably, artificial intelligence (AI) technology. This review provides an overview of these advancements, with a focus on deep learning methods for early detection of PDA.
Collapse
Affiliation(s)
| | - Yiqiu Shen
- New York University Langone Health, New York, USA
| | | | | | | | | | - Zhen Jane Wang
- University of California, San Francisco, San Francisco, USA
| | - Linda C Chu
- Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
3
|
Ponnarassery Chandran S, Santhi N. Case Study on Analysing the Early Disease Detection of Pancreatic Ductal Adenocarcinoma in Korean Association for Clinical Oncology. Am J Clin Oncol 2024; 47:475-484. [PMID: 38963000 DOI: 10.1097/coc.0000000000001118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) is the most pervasive sort of pancreatic malignant growth. Due to the lack of early symptoms and effective methods for early detection and screening, the majority of patients (80% to 85%) are diagnosed with advanced metastatic or locally advanced disease, resulting in a low 5-year survival rate of 12%. The case study represents a comprehensive investigation into the intricate landscape of pancreatic cancer diagnosis within the Korean population. METHODS Grounded in epidemiological bits of knowledge, the review plans to disentangle the particular examples, commonness, and segment attributes of PDAC in Korea. By scrutinizing current diagnostic modalities, including conventional imaging techniques, molecular markers, and emerging technologies, the research seeks to evaluate the strengths and limitations of existing approaches within the Korean clinical context. Central to the study is an exploration of the collaborative initiatives spearheaded by the Association of Clinical Oncology in Korea in the domain of PDAC early detection. Analysing research projects, clinical trials, and interdisciplinary collaborations, the case study sheds light on the association's pivotal role in driving innovation and progress in oncology. RESULTS The goal is to offer a detailed analysis of how the association helps in furthering knowledge and enhancing results in the management of PDAC. The case study delves into the implications of early PDAC detection for patient outcomes, emphasizing the significance of timely interventions and tailored treatment strategies. By outlining the potential benefits and challenges associated with early diagnosis, the study aims to inform health care policies, shape clinical guidelines, and guide future research priorities. CONCLUSION Through a holistic approach, the case study endeavours to offer important experiences into the multifaceted landscape of PDAC early detection within the Korean health care system, contributing to the broader discourse on effective oncological practices and patient care.
Collapse
Affiliation(s)
- Sijithra Ponnarassery Chandran
- Department of Electronics and Communication Engineering, Noorul Islam Centre for Higher Education, Kanyakumari District, Tamil Nadu, India
| | | |
Collapse
|
4
|
Ahmed TM, Kawamoto S, Lopez-Ramirez F, Yasrab M, Hruban RH, Fishman EK, Chu LC. Early detection of pancreatic cancer in the era of precision medicine. Abdom Radiol (NY) 2024; 49:3559-3573. [PMID: 38761272 DOI: 10.1007/s00261-024-04358-w] [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: 03/31/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/20/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related mortality and it is often diagnosed at advanced stages due to non-specific clinical presentation. Disease detection at localized disease stage followed by surgical resection remains the only potentially curative treatment. In this era of precision medicine, a multifaceted approach to early detection of PDAC includes targeted screening in high-risk populations, serum biomarkers and "liquid biopsies", and artificial intelligence augmented tumor detection from radiologic examinations. In this review, we will review these emerging techniques in the early detection of PDAC.
Collapse
Affiliation(s)
- Taha M Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Felipe Lopez-Ramirez
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Mohammad Yasrab
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Ralph H Hruban
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Linda C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA.
| |
Collapse
|
5
|
Aouad T, Laurent V, Levant P, Rode A, Brillat-Savarin N, Gaillot P, Hoeffel C, Frampas E, Barat M, Russo R, Wagner M, Zappa M, Ernst O, Delagnes A, Fillias Q, Dawi L, Savoye-Collet C, Copin P, Calame P, Reizine E, Luciani A, Bellin MF, Talbot H, Lassau N. Detection and characterization of pancreatic lesion with artificial intelligence: The SFR 2023 artificial intelligence data challenge. Diagn Interv Imaging 2024; 105:395-399. [PMID: 39048455 DOI: 10.1016/j.diii.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
PURPOSE The purpose of the 2023 SFR data challenge was to invite researchers to develop artificial intelligence (AI) models to identify the presence of a pancreatic mass and distinguish between benign and malignant pancreatic masses on abdominal computed tomography (CT) examinations. MATERIALS AND METHODS Anonymized abdominal CT examinations acquired during the portal venous phase were collected from 18 French centers. Abdominal CT examinations were divided into three groups including CT examinations with no lesion, CT examinations with benign pancreatic mass, or CT examinations with malignant pancreatic mass. Each team included at least one radiologist, one data scientist, and one engineer. Pancreatic lesions were annotated by expert radiologists. CT examinations were distributed in balanced batches via a Health Data Hosting certified platform. Data were distributed into four batches, two for training, one for internal evaluation, and one for the external evaluation. Training used 83 % of the data from 14 centers and external evaluation used data from the other four centers. The metric (i.e., final score) used to rank the participants was a weighted average of mean sensitivity, mean precision and mean area under the curve. RESULTS A total of 1037 abdominal CT examinations were divided into two training sets (including 500 and 232 CT examinations), an internal evaluation set (including 139 CT examinations), and an external evaluation set (including 166 CT examinations). The training sets were distributed on September 7 and October 13, 2023, and evaluation sets on October 15, 2023. Ten teams with a total of 93 members participated to the data challenge, with the best final score being 0.72. CONCLUSION This SFR 2023 data challenge based on multicenter CT data suggests that the use of AI for pancreatic lesions detection is possible on real data, but the distinction between benign and malignant pancreatic lesions remains challenging.
Collapse
Affiliation(s)
- Theodore Aouad
- CentraleSupelec, INRIA, CVN, Université Paris-Saclay, 91190 Gif-sur-Yvette, France.
| | - Valerie Laurent
- Department of Radiology, University Hospital of Nancy, Laboratoire IADI INSERM U 1254, 54035 Nancy, France
| | - Paul Levant
- Société Française de Radiologie, 75013 Paris, France
| | - Agnes Rode
- Department of Diagnostic and Interventional Radiology, Hospices Civils de Lyon, Hôpital de la Croix Rousse, 69317 Lyon, France
| | | | - Pénélope Gaillot
- Department of Diagnostic and Interventional Radiology, Assistance Publique-Hopitaux de Paris, CHU de Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Christine Hoeffel
- Department of Radiology, HMB, CHU Reims, 51100 Reims, France; CReSTIC, Université de Reims-Champagne-Ardenne, UFR Sciences Exactes et Naturelles, 51100 Reims, France
| | - Eric Frampas
- Department of Radiology, Hôtel Dieu, CHU Nantes, 44093 Nantes, France
| | - Maxime Barat
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hopitaux de Paris, 75014 Paris, France; Génomique et Signalisation des Tumeurs Endocrines, Institut Cochin, INSERM U 1016, CNRS UMR8104, 75014 Paris, France; Faculté de Médecine, Université Paris Cité, 75006 Paris, France
| | - Roberta Russo
- Department of Radiology, Hôpital Paul Brousse, Assistance Publique-Hopitaux de Paris, 94800 Villejuif, France
| | - Mathilde Wagner
- Department of Radiology, Assistance Publique-Hopitaux de Paris, Sorbonne Université, Hôpital Universitaire Pitié-Salpêtrière, 75013 Paris, France
| | - Magaly Zappa
- Department of Radiology, Centre Hospitalier de Cayenne, Cayenne 97306, France
| | - Olivier Ernst
- Medical Imaging Department, Lille University Hospital, 59000 Lille, France
| | - Anais Delagnes
- Department of Radiology, CHU Angers, Angers University Hospital, 49933 Angers, France
| | - Quentin Fillias
- Department of Radiology, Hospital Lapeyronie, CHU Montpellier, 34000 Montpellier, France
| | - Lama Dawi
- Department of Radiology, Gustave Roussy, 94805 Villejuif, France
| | - Céline Savoye-Collet
- Department of Radiology, Normandie Université, UNIROUEN, Quantif-LITIS EA 4108, Rouen University Hospital, 76031 Rouen, France
| | - Pauline Copin
- Department of Radiology, Hôpital Beaujon, AP-HP.Nord, 92110 Clichy, France
| | - Paul Calame
- Department of Radiology, University of Bourgogne Franche-Comté, CHU Besançon, 25030 Besançon, France
| | - Edouard Reizine
- Department of Radiology, Hopital Henri Mondor, Assistance Publique-Hopitaux de Paris, University Paris Est Créteil, 94000 Créteil, France
| | - Alain Luciani
- Société Française de Radiologie, 75013 Paris, France; Department of Radiology, Hopital Henri Mondor, Assistance Publique-Hopitaux de Paris, University Paris Est Créteil, 94000 Créteil, France; INSERM, U955, Team 18, 94000 Créteil, France
| | - Marie-France Bellin
- Société Française de Radiologie, 75013 Paris, France; Department of Diagnostic and Interventional Radiology, Assistance Publique-Hopitaux de Paris, CHU de Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Hugues Talbot
- CentraleSupelec, INRIA, CVN, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Nathalie Lassau
- Department of Radiology, Gustave Roussy, 94805 Villejuif, France; Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Inserm, CNRS, CEA, BIOMAPS, UMR 1281, Université Paris-Saclay, Villejuif 94800, France
| |
Collapse
|
6
|
Zhang G, Gao Q, Zhan Q, Wang L, Song B, Chen Y, Bian Y, Ma C, Lu J, Shao C. Label-free differentiation of pancreatic pathologies from normal pancreas utilizing end-to-end three-dimensional multimodal networks on CT. Clin Radiol 2024; 79:e1159-e1166. [PMID: 38969545 DOI: 10.1016/j.crad.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/10/2024] [Accepted: 06/05/2024] [Indexed: 07/07/2024]
Abstract
AIMS To investigate the utilization of an end-to-end multimodal convolutional model in the rapid and accurate diagnosis of pancreatic diseases using abdominal CT images. MATERIALS AND METHODS In this study, a novel lightweight label-free end-to-end multimodal network (eeMulNet) model was proposed for the rapid and precise diagnosis of abnormal pancreas. The eeMulNet consists of two steps: pancreatic region localization and multimodal CT diagnosis integrating textual and image data. A research dataset comprising 715 CT scans with various types of pancreas diseases and 228 CT scans from a control group was collected. The training set and independent test set for the multimodal classification network were randomly divided in an 8:2 ratio (755 for training and 188 for testing). RESULTS The eeMulNet model demonstrated outstanding performance on an independent test set of 188 CT scans (Normal: 45, Abnormal: 143), with an area under the curve (AUC) of 1.0, accuracy of 100%, and sensitivity of 100%. The average testing duration per patient was 41.04 seconds, while the classification network took only 0.04 seconds. CONCLUSIONS The proposed eeMulNet model offers a promising approach for the diagnosis of pancreatic diseases. It can support the identification of suspicious cases during daily radiology work and enhance the accuracy of pancreatic disease diagnosis. The codes and models of eeMulNet are publicly available at Rudeguy1/eeMulNet (github.com).
Collapse
Affiliation(s)
- G Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai 200433, China.
| | - Q Gao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai 200433, China.
| | - Q Zhan
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai 200433, China.
| | - L Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - B Song
- Department of Pancreatic Surgery, Changhai Hospital of Shanghai, Naval Medical University, Shanghai 200433, China.
| | - Y Chen
- College of Electronic and Information Engineering, Tongji University, Shanghai 201804, China.
| | - Y Bian
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai 200433, China.
| | - C Ma
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai 200433, China; College of Electronic and Information Engineering, Tongji University, Shanghai 201804, China.
| | - J Lu
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai 200433, China.
| | - C Shao
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
7
|
Alzubaidi L, Al-Dulaimi K, Salhi A, Alammar Z, Fadhel MA, Albahri AS, Alamoodi AH, Albahri OS, Hasan AF, Bai J, Gilliland L, Peng J, Branni M, Shuker T, Cutbush K, Santamaría J, Moreira C, Ouyang C, Duan Y, Manoufali M, Jomaa M, Gupta A, Abbosh A, Gu Y. Comprehensive review of deep learning in orthopaedics: Applications, challenges, trustworthiness, and fusion. Artif Intell Med 2024; 155:102935. [PMID: 39079201 DOI: 10.1016/j.artmed.2024.102935] [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: 06/01/2023] [Revised: 03/18/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024]
Abstract
Deep learning (DL) in orthopaedics has gained significant attention in recent years. Previous studies have shown that DL can be applied to a wide variety of orthopaedic tasks, including fracture detection, bone tumour diagnosis, implant recognition, and evaluation of osteoarthritis severity. The utilisation of DL is expected to increase, owing to its ability to present accurate diagnoses more efficiently than traditional methods in many scenarios. This reduces the time and cost of diagnosis for patients and orthopaedic surgeons. To our knowledge, no exclusive study has comprehensively reviewed all aspects of DL currently used in orthopaedic practice. This review addresses this knowledge gap using articles from Science Direct, Scopus, IEEE Xplore, and Web of Science between 2017 and 2023. The authors begin with the motivation for using DL in orthopaedics, including its ability to enhance diagnosis and treatment planning. The review then covers various applications of DL in orthopaedics, including fracture detection, detection of supraspinatus tears using MRI, osteoarthritis, prediction of types of arthroplasty implants, bone age assessment, and detection of joint-specific soft tissue disease. We also examine the challenges for implementing DL in orthopaedics, including the scarcity of data to train DL and the lack of interpretability, as well as possible solutions to these common pitfalls. Our work highlights the requirements to achieve trustworthiness in the outcomes generated by DL, including the need for accuracy, explainability, and fairness in the DL models. We pay particular attention to fusion techniques as one of the ways to increase trustworthiness, which have also been used to address the common multimodality in orthopaedics. Finally, we have reviewed the approval requirements set forth by the US Food and Drug Administration to enable the use of DL applications. As such, we aim to have this review function as a guide for researchers to develop a reliable DL application for orthopaedic tasks from scratch for use in the market.
Collapse
Affiliation(s)
- Laith Alzubaidi
- School of Mechanical, Medical, and Process Engineering, Queensland University of Technology, Brisbane, QLD 4000, Australia; QUASR/ARC Industrial Transformation Training Centre-Joint Biomechanics, Queensland University of Technology, Brisbane, QLD 4000, Australia; Research and Development department, Akunah Med Technology Pty Ltd Co, Brisbane, QLD 4120, Australia.
| | - Khamael Al-Dulaimi
- Computer Science Department, College of Science, Al-Nahrain University, Baghdad, Baghdad 10011, Iraq; School of Electrical Engineering and Robotics, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Asma Salhi
- QUASR/ARC Industrial Transformation Training Centre-Joint Biomechanics, Queensland University of Technology, Brisbane, QLD 4000, Australia; Research and Development department, Akunah Med Technology Pty Ltd Co, Brisbane, QLD 4120, Australia
| | - Zaenab Alammar
- School of Computer Science, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Mohammed A Fadhel
- Research and Development department, Akunah Med Technology Pty Ltd Co, Brisbane, QLD 4120, Australia
| | - A S Albahri
- Technical College, Imam Ja'afar Al-Sadiq University, Baghdad, Iraq
| | - A H Alamoodi
- Institute of Informatics and Computing in Energy, Universiti Tenaga Nasional, Kajang 43000, Malaysia
| | - O S Albahri
- Australian Technical and Management College, Melbourne, Australia
| | - Amjad F Hasan
- Faculty of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| | - Jinshuai Bai
- School of Mechanical, Medical, and Process Engineering, Queensland University of Technology, Brisbane, QLD 4000, Australia; QUASR/ARC Industrial Transformation Training Centre-Joint Biomechanics, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Luke Gilliland
- QUASR/ARC Industrial Transformation Training Centre-Joint Biomechanics, Queensland University of Technology, Brisbane, QLD 4000, Australia; Research and Development department, Akunah Med Technology Pty Ltd Co, Brisbane, QLD 4120, Australia
| | - Jing Peng
- Research and Development department, Akunah Med Technology Pty Ltd Co, Brisbane, QLD 4120, Australia
| | - Marco Branni
- QUASR/ARC Industrial Transformation Training Centre-Joint Biomechanics, Queensland University of Technology, Brisbane, QLD 4000, Australia; Research and Development department, Akunah Med Technology Pty Ltd Co, Brisbane, QLD 4120, Australia
| | - Tristan Shuker
- QUASR/ARC Industrial Transformation Training Centre-Joint Biomechanics, Queensland University of Technology, Brisbane, QLD 4000, Australia; St Andrew's War Memorial Hospital, Brisbane, QLD 4000, Australia
| | - Kenneth Cutbush
- QUASR/ARC Industrial Transformation Training Centre-Joint Biomechanics, Queensland University of Technology, Brisbane, QLD 4000, Australia; St Andrew's War Memorial Hospital, Brisbane, QLD 4000, Australia
| | - Jose Santamaría
- Department of Computer Science, University of Jaén, Jaén 23071, Spain
| | - Catarina Moreira
- Data Science Institute, University of Technology Sydney, Australia
| | - Chun Ouyang
- School of Information Systems, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Ye Duan
- School of Computing, Clemson University, Clemson, 29631, SC, USA
| | - Mohamed Manoufali
- CSIRO, Kensington, WA 6151, Australia; School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, QLD 4067, Australia
| | - Mohammad Jomaa
- QUASR/ARC Industrial Transformation Training Centre-Joint Biomechanics, Queensland University of Technology, Brisbane, QLD 4000, Australia; St Andrew's War Memorial Hospital, Brisbane, QLD 4000, Australia
| | - Ashish Gupta
- School of Mechanical, Medical, and Process Engineering, Queensland University of Technology, Brisbane, QLD 4000, Australia; QUASR/ARC Industrial Transformation Training Centre-Joint Biomechanics, Queensland University of Technology, Brisbane, QLD 4000, Australia; Research and Development department, Akunah Med Technology Pty Ltd Co, Brisbane, QLD 4120, Australia
| | - Amin Abbosh
- School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, QLD 4067, Australia
| | - Yuantong Gu
- School of Mechanical, Medical, and Process Engineering, Queensland University of Technology, Brisbane, QLD 4000, Australia; QUASR/ARC Industrial Transformation Training Centre-Joint Biomechanics, Queensland University of Technology, Brisbane, QLD 4000, Australia
| |
Collapse
|
8
|
Marcinkiewicz AM, Buchwald M, Shanbhag A, Bednarski BP, Killekar A, Miller RJ, Builoff V, Lemley M, Berman DS, Dey D, Slomka PJ, Weintraub E. AI for Multistructure Incidental Findings and Mortality Prediction at Chest CT in Lung Cancer Screening. Radiology 2024; 312:e240541. [PMID: 39287522 PMCID: PMC11427857 DOI: 10.1148/radiol.240541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 09/19/2024]
Abstract
Background Incidental extrapulmonary findings are commonly detected on chest CT scans and can be clinically important. Purpose To integrate artificial intelligence (AI)-based segmentation for multiple structures, coronary artery calcium (CAC), and epicardial adipose tissue with automated feature extraction methods and machine learning to detect extrapulmonary abnormalities and predict all-cause mortality (ACM) in a large multicenter cohort. Materials and Methods In this post hoc analysis, baseline chest CT scans in patients enrolled in the National Lung Screening Trial (NLST) from August 2002 to September 2007 were included from 33 participating sites. Per scan, 32 structures were segmented with a multistructure model. For each structure, 15 clinically interpretable radiomic features were quantified. Four general codes describing abnormalities reported by NLST radiologists were applied to identify extrapulmonary significant incidental findings on the CT scans. Death at 2-year and 10-year follow-up and the presence of extrapulmonary significant incidental findings were predicted with ensemble AI models, and individualized structure risk scores were evaluated. Area under the receiver operating characteristic curve (AUC) analysis was used to evaluate the performance of the models for prediction of ACM and extrapulmonary significant incidental findings. The Pearson χ2 test and Kruskal-Wallis rank sum test were used for statistical analyses. Results A total of 24 401 participants (median age, 61 years [IQR, 57-65 years]; 14 468 male) were included. In 3880 of 24 401 participants (16%), 4283 extrapulmonary significant incidental findings were reported. During the 10-year follow-up, 3389 of 24 401 participants (14%) died. CAC had the highest feature importance for predicting the three study end points. The 10-year ACM model demonstrated the best AUC performance (0.72; per-year mortality of 2.6% above and 0.8% below the risk threshold), followed by 2-year ACM (0.71; per-year mortality of 1.13% above and 0.3% below the risk threshold) and prediction of extrapulmonary significant incidental findings (0.70; probability of occurrence of 25.4% above and 9.6% below the threshold). Conclusion A fully automated AI model indicated extrapulmonary structures at risk on chest CT scans and predicted ACM with explanations. ClinicalTrials.gov Identifier: NCT00047385 © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Yanagawa and Hata in this issue.
Collapse
Affiliation(s)
- Anna M. Marcinkiewicz
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| | - Mikolaj Buchwald
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| | - Aakash Shanbhag
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| | - Bryan P. Bednarski
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| | - Aditya Killekar
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| | - Robert J.H. Miller
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| | - Valerie Builoff
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| | - Mark Lemley
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| | - Daniel S. Berman
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| | - Damini Dey
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| | - Piotr J. Slomka
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| | - Elizabeth Weintraub
- From the Departments of Medicine, Division of Artificial Intelligence
in Medicine, Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, 6500
Wilshire Blvd, Los Angeles, CA 90048 (A.M.M., M.B., A.S., B.P.B., A.K.,
R.J.H.M., V.B., M.L., D.S.B., D.D., P.J.S.); Signal and Image Processing
Institute, Ming Hsieh Department of Electrical and Computer Engineering,
University of Southern California, Los Angeles, Calif (A.S.); and Department of
Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
(R.J.H.M.)
| |
Collapse
|
9
|
Liu W, Zhang B, Liu T, Jiang J, Liu Y. Artificial Intelligence in Pancreatic Image Analysis: A Review. SENSORS (BASEL, SWITZERLAND) 2024; 24:4749. [PMID: 39066145 PMCID: PMC11280964 DOI: 10.3390/s24144749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Pancreatic cancer is a highly lethal disease with a poor prognosis. Its early diagnosis and accurate treatment mainly rely on medical imaging, so accurate medical image analysis is especially vital for pancreatic cancer patients. However, medical image analysis of pancreatic cancer is facing challenges due to ambiguous symptoms, high misdiagnosis rates, and significant financial costs. Artificial intelligence (AI) offers a promising solution by relieving medical personnel's workload, improving clinical decision-making, and reducing patient costs. This study focuses on AI applications such as segmentation, classification, object detection, and prognosis prediction across five types of medical imaging: CT, MRI, EUS, PET, and pathological images, as well as integrating these imaging modalities to boost diagnostic accuracy and treatment efficiency. In addition, this study discusses current hot topics and future directions aimed at overcoming the challenges in AI-enabled automated pancreatic cancer diagnosis algorithms.
Collapse
Affiliation(s)
- Weixuan Liu
- Sydney Smart Technology College, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China; (W.L.); (B.Z.)
| | - Bairui Zhang
- Sydney Smart Technology College, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China; (W.L.); (B.Z.)
| | - Tao Liu
- School of Mathematics and Statistics, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China;
| | - Juntao Jiang
- College of Control Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yong Liu
- College of Control Science and Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
10
|
Maurya R, Chug I, Vudatha V, Palma AM. Applications of spatial transcriptomics and artificial intelligence to develop integrated management of pancreatic cancer. Adv Cancer Res 2024; 163:107-136. [PMID: 39271261 DOI: 10.1016/bs.acr.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Cancer is a complex disease intrinsically associated with cellular processes and gene expression. With the development of techniques such as single-cell sequencing and sequential fluorescence in situ hybridization (seqFISH), it was possible to map the location of cells based on their gene expression with more precision. Moreover, in recent years, many tools have been developed to analyze these extensive datasets by integrating machine learning and artificial intelligence in a comprehensive manner. Since these tools analyze sequencing data, they offer the chance to analyze any tissue regardless of its origin. By applying this to cancer settings, spatial transcriptomic analysis based on artificial intelligence may help us understand cell-cell communications within the tumor microenvironment. Another advantage of this analysis is the identification of new biomarkers and therapeutic targets. The integration of such analysis with other omics data and with routine exams such as magnetic resonance imaging can help physicians with the earlier diagnosis of tumors as well as establish a more personalized treatment for pancreatic cancer patients. In this review, we give an overview description of pancreatic cancer, describe how spatial transcriptomics and artificial intelligence have been used to study pancreatic cancer and provide examples of how integrating these tools may help physicians manage pancreatic cancer in a more personalized approach.
Collapse
Affiliation(s)
- Rishabh Maurya
- Department of Surgery, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Isha Chug
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Vignesh Vudatha
- Department of Surgery, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - António M Palma
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States; VCU Institute of Molecular Medicine, Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
11
|
Ramaekers M, Viviers CGA, Hellström TAE, Ewals LJS, Tasios N, Jacobs I, Nederend J, Sommen FVD, Luyer MDP. Improved Pancreatic Cancer Detection and Localization on CT Scans: A Computer-Aided Detection Model Utilizing Secondary Features. Cancers (Basel) 2024; 16:2403. [PMID: 39001465 PMCID: PMC11240790 DOI: 10.3390/cancers16132403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
The early detection of pancreatic ductal adenocarcinoma (PDAC) is essential for optimal treatment of pancreatic cancer patients. We propose a tumor detection framework to improve the detection of pancreatic head tumors on CT scans. In this retrospective research study, CT images of 99 patients with pancreatic head cancer and 98 control cases from the Catharina Hospital Eindhoven were collected. A multi-stage 3D U-Net-based approach was used for PDAC detection including clinically significant secondary features such as pancreatic duct and common bile duct dilation. The developed algorithm was evaluated using a local test set comprising 59 CT scans. The model was externally validated in 28 pancreatic cancer cases of a publicly available medical decathlon dataset. The tumor detection framework achieved a sensitivity of 0.97 and a specificity of 1.00, with an area under the receiver operating curve (AUROC) of 0.99, in detecting pancreatic head cancer in the local test set. In the external test set, we obtained similar results, with a sensitivity of 1.00. The model provided the tumor location with acceptable accuracy obtaining a DICE Similarity Coefficient (DSC) of 0.37. This study shows that a tumor detection framework utilizing CT scans and secondary signs of pancreatic cancer can detect pancreatic tumors with high accuracy.
Collapse
Affiliation(s)
- Mark Ramaekers
- Department of Surgery, Catharina Cancer Institute, Catharina Hospital Eindhoven, EJ 5623 Eindhoven, The Netherlands
| | - Christiaan G A Viviers
- Department of Electrical Engineering, Eindhoven University of Technology, AZ 5612 Eindhoven, The Netherlands
| | - Terese A E Hellström
- Department of Electrical Engineering, Eindhoven University of Technology, AZ 5612 Eindhoven, The Netherlands
| | - Lotte J S Ewals
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, EJ 5623 Eindhoven, The Netherlands
| | - Nick Tasios
- Department of Hospital Services and Informatics, Philips Research, AE 5656 Eindhoven, The Netherlands
| | - Igor Jacobs
- Department of Hospital Services and Informatics, Philips Research, AE 5656 Eindhoven, The Netherlands
| | - Joost Nederend
- Department of Radiology, Catharina Cancer Institute, Catharina Hospital Eindhoven, EJ 5623 Eindhoven, The Netherlands
| | - Fons van der Sommen
- Department of Electrical Engineering, Eindhoven University of Technology, AZ 5612 Eindhoven, The Netherlands
| | - Misha D P Luyer
- Department of Surgery, Catharina Cancer Institute, Catharina Hospital Eindhoven, EJ 5623 Eindhoven, The Netherlands
| |
Collapse
|
12
|
Kahraman G, Haberal KM, Dilek ON. Imaging features and management of focal liver lesions. World J Radiol 2024; 16:139-167. [PMID: 38983841 PMCID: PMC11229941 DOI: 10.4329/wjr.v16.i6.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/28/2024] [Accepted: 05/22/2024] [Indexed: 06/26/2024] Open
Abstract
Notably, the number of incidentally detected focal liver lesions (FLLs) has increased dramatically in recent years due to the increased use of radiological imaging. The diagnosis of FLLs can be made through a well-documented medical history, physical examination, laboratory tests, and appropriate imaging methods. Although benign FLLs are more common than malignant ones in adults, even in patients with primary malignancy, accurate diagnosis of incidental FLLs is of utmost clinical significance. In clinical practice, FLLs are frequently evaluated non-invasively using ultrasound (US), computed tomography (CT), and magnetic resonance imaging (MRI). Although US is a cost-effective and widely used imaging method, its diagnostic specificity and sensitivity for FLL characterization are limited. FLLs are primarily characterized by obtaining enhancement patterns through dynamic contrast-enhanced CT and MRI. MRI is a problem-solving method with high specificity and sensitivity, commonly used for the evaluation of FLLs that cannot be characterized by US or CT. Recent technical advancements in MRI, along with the use of hepatobiliary-specific MRI contrast agents, have significantly improved the success of FLL characterization and reduced unnecessary biopsies. The American College of Radiology (ACR) appropriateness criteria are evidence-based recommendations intended to assist clinicians in selecting the optimal imaging or treatment option for their patients. ACR Appropriateness Criteria Liver Lesion-Initial Characterization guideline provides recommendations for the imaging methods that should be used for the characterization of incidentally detected FLLs in various clinical scenarios. The American College of Gastroenterology (ACG) Clinical Guideline offers evidence-based recommendations for both the diagnosis and management of FLL. American Association for the Study of Liver Diseases (AASLD) Practice Guidance provides an approach to the diagnosis and management of patients with hepatocellular carcinoma. In this article, FLLs are reviewed with a comprehensive analysis of ACR Appropriateness Criteria, ACG Clinical Guideline, AASLD Practice Guidance, and current medical literature from peer-reviewed journals. The article includes a discussion of imaging methods used for the assessment of FLL, current recommended imaging techniques, innovations in liver imaging, contrast agents, imaging features of common nonmetastatic benign and malignant FLL, as well as current management recommendations.
Collapse
Affiliation(s)
- Gökhan Kahraman
- Department of Radiology, Suluova State Hospital, Amasya 05500, Türkiye
| | - Kemal Murat Haberal
- Department of Radiology, Başkent University Faculty of Medicine, Ankara 06490, Türkiye
| | - Osman Nuri Dilek
- Department of Surgery, İzmir Katip Celebi University, School of Medicine, İzmir 35150, Türkiye
| |
Collapse
|
13
|
Mukund A, Afridi MA, Karolak A, Park MA, Permuth JB, Rasool G. Pancreatic Ductal Adenocarcinoma (PDAC): A Review of Recent Advancements Enabled by Artificial Intelligence. Cancers (Basel) 2024; 16:2240. [PMID: 38927945 PMCID: PMC11201559 DOI: 10.3390/cancers16122240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) remains one of the most formidable challenges in oncology, characterized by its late detection and poor prognosis. Artificial intelligence (AI) and machine learning (ML) are emerging as pivotal tools in revolutionizing PDAC care across various dimensions. Consequently, many studies have focused on using AI to improve the standard of PDAC care. This review article attempts to consolidate the literature from the past five years to identify high-impact, novel, and meaningful studies focusing on their transformative potential in PDAC management. Our analysis spans a broad spectrum of applications, including but not limited to patient risk stratification, early detection, and prediction of treatment outcomes, thereby highlighting AI's potential role in enhancing the quality and precision of PDAC care. By categorizing the literature into discrete sections reflective of a patient's journey from screening and diagnosis through treatment and survivorship, this review offers a comprehensive examination of AI-driven methodologies in addressing the multifaceted challenges of PDAC. Each study is summarized by explaining the dataset, ML model, evaluation metrics, and impact the study has on improving PDAC-related outcomes. We also discuss prevailing obstacles and limitations inherent in the application of AI within the PDAC context, offering insightful perspectives on potential future directions and innovations.
Collapse
Affiliation(s)
- Ashwin Mukund
- Department of Machine Learning, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (A.M.); (A.K.)
| | - Muhammad Ali Afridi
- School of Electrical Engineering and Computer Science (SEECS), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan;
| | - Aleksandra Karolak
- Department of Machine Learning, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (A.M.); (A.K.)
| | - Margaret A. Park
- Departments of Cancer Epidemiology and Gastrointestinal Oncology, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (M.A.P.); (J.B.P.)
| | - Jennifer B. Permuth
- Departments of Cancer Epidemiology and Gastrointestinal Oncology, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (M.A.P.); (J.B.P.)
| | - Ghulam Rasool
- Department of Machine Learning, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (A.M.); (A.K.)
| |
Collapse
|
14
|
Miao Q, Wang X, Cui J, Zheng H, Xie Y, Zhu K, Chai R, Jiang Y, Feng D, Zhang X, Shi F, Tan X, Fan G, Liang K. Artificial intelligence to predict T4 stage of pancreatic ductal adenocarcinoma using CT imaging. Comput Biol Med 2024; 171:108125. [PMID: 38340439 DOI: 10.1016/j.compbiomed.2024.108125] [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: 10/30/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND The accurate assessment of T4 stage of pancreatic ductal adenocarcinoma (PDAC) has consistently presented a considerable difficulty for radiologists. This study aimed to develop and validate an automated artificial intelligence (AI) pipeline for the prediction of T4 stage of PDAC using contrast-enhanced CT imaging. METHODS The data were obtained retrospectively from consecutive patients with surgically resected and pathologically proved PDAC at two institutions between July 2017 and June 2022. Initially, a deep learning (DL) model was developed to segment PDAC. Subsequently, radiomics features were extracted from the automatically segmented region of interest (ROI), which encompassed both the tumor region and a 3 mm surrounding area, to construct a predictive model for determining T4 stage of PDAC. The assessment of the models' performance involved the calculation of the area under the receiver operating characteristic curve (AUC), sensitivity, and specificity. RESULTS The study encompassed a cohort of 509 PDAC patients, with a median age of 62 years (interquartile range: 55-67). The proportion of patients in T4 stage within the model was 16.9%. The model achieved an AUC of 0.849 (95% CI: 0.753-0.940), a sensitivity of 0.875, and a specificity of 0.728 in predicting T4 stage of PDAC. The performance of the model was determined to be comparable to that of two experienced abdominal radiologists (AUCs: 0.849 vs. 0.834 and 0.857). CONCLUSION The automated AI pipeline utilizing tumor and peritumor-related radiomics features demonstrated comparable performance to that of senior abdominal radiologists in predicting T4 stage of PDAC.
Collapse
Affiliation(s)
- Qi Miao
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Xuechun Wang
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Jingjing Cui
- Department of Research and Development, United Imaging Intelligence (Beijing) Co., Ltd., Bejing, China
| | - Haoxin Zheng
- Department of Computer Science, University of California, Los Angeles, USA
| | - Yan Xie
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Kexin Zhu
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Ruimei Chai
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Yuanxi Jiang
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Dongli Feng
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Xin Zhang
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Feng Shi
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Xiaodong Tan
- Department of General Surgery/Pancreatic and Thyroid Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guoguang Fan
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China.
| | - Keke Liang
- Department of General Surgery/Pancreatic and Thyroid Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
15
|
Scherübl H. [Early detection of sporadic pancreatic cancer]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:412-419. [PMID: 37827502 DOI: 10.1055/a-2114-9847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The incidence of pancreatic cancer is rising. At present, pancreatic cancer is the third most common cancer-causing death in Germany, but it is expected to become the second in 2030 and finally the leading cause of cancer death in 2050. Pancreatic ductal adenocarcinoma (PC) is generally diagnosed at advanced stages, and 5-year-survival has remained poor. Early detection of sporadic PC at stage IA, however, can yield a 5-year-survival rate of about 80%. Early detection initiatives aim at identifying persons at high risk. People with new-onset diabetes at age 50 or older have attracted much interest. Novel strategies regarding how to detect sporadic PC at an early stage are being discussed.
Collapse
Affiliation(s)
- Hans Scherübl
- Klinik für Innere Medizin; Gastroenterol., GI Onkol. u. Infektiol., Vivantes Klinikum Am Urban, Berlin, Germany
- Akademisches Lehrkrankenhaus der Charité, Berlin, Germany
| |
Collapse
|
16
|
Niu Z, O'Farrell A, Li J, Reffsin S, Jain N, Dardani I, Goyal Y, Raj A. Piscis: a novel loss estimator of the F1 score enables accurate spot detection in fluorescence microscopy images via deep learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578123. [PMID: 38352551 PMCID: PMC10862914 DOI: 10.1101/2024.01.31.578123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Single-molecule RNA fluorescence in situ hybridization (RNA FISH)-based spatial transcriptomics methods have enabled the accurate quantification of gene expression at single-cell resolution by visualizing transcripts as diffraction-limited spots. While these methods generally scale to large samples, image analysis remains challenging, often requiring manual parameter tuning. We present Piscis, a fully automatic deep learning algorithm for spot detection trained using a novel loss function, the SmoothF1 loss, that approximates the F1 score to directly penalize false positives and false negatives but remains differentiable and hence usable for training by deep learning approaches. Piscis was trained and tested on a diverse dataset composed of 358 manually annotated experimental RNA FISH images representing multiple cell types and 240 additional synthetic images. Piscis outperforms other state-of-the-art spot detection methods, enabling accurate, high-throughput analysis of RNA FISH-derived imaging data without the need for manual parameter tuning.
Collapse
Affiliation(s)
- Zijian Niu
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physics and Astronomy, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Aoife O'Farrell
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingxin Li
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sam Reffsin
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Naveen Jain
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ian Dardani
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Yogesh Goyal
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Arjun Raj
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
17
|
Tripathi S, Tabari A, Mansur A, Dabbara H, Bridge CP, Daye D. From Machine Learning to Patient Outcomes: A Comprehensive Review of AI in Pancreatic Cancer. Diagnostics (Basel) 2024; 14:174. [PMID: 38248051 PMCID: PMC10814554 DOI: 10.3390/diagnostics14020174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Pancreatic cancer is a highly aggressive and difficult-to-detect cancer with a poor prognosis. Late diagnosis is common due to a lack of early symptoms, specific markers, and the challenging location of the pancreas. Imaging technologies have improved diagnosis, but there is still room for improvement in standardizing guidelines. Biopsies and histopathological analysis are challenging due to tumor heterogeneity. Artificial Intelligence (AI) revolutionizes healthcare by improving diagnosis, treatment, and patient care. AI algorithms can analyze medical images with precision, aiding in early disease detection. AI also plays a role in personalized medicine by analyzing patient data to tailor treatment plans. It streamlines administrative tasks, such as medical coding and documentation, and provides patient assistance through AI chatbots. However, challenges include data privacy, security, and ethical considerations. This review article focuses on the potential of AI in transforming pancreatic cancer care, offering improved diagnostics, personalized treatments, and operational efficiency, leading to better patient outcomes.
Collapse
Affiliation(s)
- Satvik Tripathi
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Azadeh Tabari
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Arian Mansur
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Harika Dabbara
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Christopher P. Bridge
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Dania Daye
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
18
|
Rahman A, Debnath T, Kundu D, Khan MSI, Aishi AA, Sazzad S, Sayduzzaman M, Band SS. Machine learning and deep learning-based approach in smart healthcare: Recent advances, applications, challenges and opportunities. AIMS Public Health 2024; 11:58-109. [PMID: 38617415 PMCID: PMC11007421 DOI: 10.3934/publichealth.2024004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/18/2023] [Indexed: 04/16/2024] Open
Abstract
In recent years, machine learning (ML) and deep learning (DL) have been the leading approaches to solving various challenges, such as disease predictions, drug discovery, medical image analysis, etc., in intelligent healthcare applications. Further, given the current progress in the fields of ML and DL, there exists the promising potential for both to provide support in the realm of healthcare. This study offered an exhaustive survey on ML and DL for the healthcare system, concentrating on vital state of the art features, integration benefits, applications, prospects and future guidelines. To conduct the research, we found the most prominent journal and conference databases using distinct keywords to discover scholarly consequences. First, we furnished the most current along with cutting-edge progress in ML-DL-based analysis in smart healthcare in a compendious manner. Next, we integrated the advancement of various services for ML and DL, including ML-healthcare, DL-healthcare, and ML-DL-healthcare. We then offered ML and DL-based applications in the healthcare industry. Eventually, we emphasized the research disputes and recommendations for further studies based on our observations.
Collapse
Affiliation(s)
- Anichur Rahman
- Department of CSE, National Institute of Textile Engineering and Research (NITER), Constituent Institute of the University of Dhaka, Savar, Dhaka-1350
- Department of CSE, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Tanoy Debnath
- Department of CSE, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
- Department of CSE, Green University of Bangladesh, 220/D, Begum Rokeya Sarani, Dhaka -1207, Bangladesh
| | - Dipanjali Kundu
- Department of CSE, National Institute of Textile Engineering and Research (NITER), Constituent Institute of the University of Dhaka, Savar, Dhaka-1350
| | - Md. Saikat Islam Khan
- Department of CSE, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Airin Afroj Aishi
- Department of Computing and Information System, Daffodil International University, Savar, Dhaka, Bangladesh
| | - Sadia Sazzad
- Department of CSE, National Institute of Textile Engineering and Research (NITER), Constituent Institute of the University of Dhaka, Savar, Dhaka-1350
| | - Mohammad Sayduzzaman
- Department of CSE, National Institute of Textile Engineering and Research (NITER), Constituent Institute of the University of Dhaka, Savar, Dhaka-1350
| | - Shahab S. Band
- Department of Information Management, International Graduate School of Artificial Intelligence, National Yunlin University of Science and Technology, Taiwan
| |
Collapse
|
19
|
Rawlani P, Ghosh NK, Kumar A. Role of artificial intelligence in the characterization of indeterminate pancreatic head mass and its usefulness in preoperative diagnosis. Artif Intell Gastroenterol 2023; 4:48-63. [DOI: 10.35712/aig.v4.i3.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/11/2023] [Accepted: 10/08/2023] [Indexed: 12/07/2023] Open
Abstract
Artificial intelligence (AI) has been used in various fields of day-to-day life and its role in medicine is immense. Understanding of oncology has been improved with the introduction of AI which helps in diagnosis, treatment planning, management, prognosis, and follow-up. It also helps to identify high-risk groups who can be subjected to timely screening for early detection of malignant conditions. It is more important in pancreatic cancer as it is one of the major causes of cancer-related deaths worldwide and there are no specific early features (clinical and radiological) for diagnosis. With improvement in imaging modalities (computed tomography, magnetic resonance imaging, endoscopic ultrasound), most often clinicians were being challenged with lesions that were difficult to diagnose with human competence. AI has been used in various other branches of medicine to differentiate such indeterminate lesions including the thyroid gland, breast, lungs, liver, adrenal gland, kidney, etc. In the case of pancreatic cancer, the role of AI has been explored and is still ongoing. This review article will focus on how AI can be used to diagnose pancreatic cancer early or differentiate it from benign pancreatic lesions, therefore, management can be planned at an earlier stage.
Collapse
Affiliation(s)
- Palash Rawlani
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| | - Nalini Kanta Ghosh
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| | - Ashok Kumar
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| |
Collapse
|
20
|
Korfiatis P, Suman G, Patnam NG, Trivedi KH, Karbhari A, Mukherjee S, Cook C, Klug JR, Patra A, Khasawneh H, Rajamohan N, Fletcher JG, Truty MJ, Majumder S, Bolan CW, Sandrasegaran K, Chari ST, Goenka AH. Automated Artificial Intelligence Model Trained on a Large Data Set Can Detect Pancreas Cancer on Diagnostic Computed Tomography Scans As Well As Visually Occult Preinvasive Cancer on Prediagnostic Computed Tomography Scans. Gastroenterology 2023; 165:1533-1546.e4. [PMID: 37657758 PMCID: PMC10843414 DOI: 10.1053/j.gastro.2023.08.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND & AIMS The aims of our case-control study were (1) to develop an automated 3-dimensional (3D) Convolutional Neural Network (CNN) for detection of pancreatic ductal adenocarcinoma (PDA) on diagnostic computed tomography scans (CTs), (2) evaluate its generalizability on multi-institutional public data sets, (3) its utility as a potential screening tool using a simulated cohort with high pretest probability, and (4) its ability to detect visually occult preinvasive cancer on prediagnostic CTs. METHODS A 3D-CNN classification system was trained using algorithmically generated bounding boxes and pancreatic masks on a curated data set of 696 portal phase diagnostic CTs with PDA and 1080 control images with a nonneoplastic pancreas. The model was evaluated on (1) an intramural hold-out test subset (409 CTs with PDA, 829 controls); (2) a simulated cohort with a case-control distribution that matched the risk of PDA in glycemically defined new-onset diabetes, and Enriching New-Onset Diabetes for Pancreatic Cancer score ≥3; (3) multi-institutional public data sets (194 CTs with PDA, 80 controls), and (4) a cohort of 100 prediagnostic CTs (i.e., CTs incidentally acquired 3-36 months before clinical diagnosis of PDA) without a focal mass, and 134 controls. RESULTS Of the CTs in the intramural test subset, 798 (64%) were from other hospitals. The model correctly classified 360 CTs (88%) with PDA and 783 control CTs (94%), with a mean accuracy 0.92 (95% CI, 0.91-0.94), area under the receiver operating characteristic (AUROC) curve of 0.97 (95% CI, 0.96-0.98), sensitivity of 0.88 (95% CI, 0.85-0.91), and specificity of 0.95 (95% CI, 0.93-0.96). Activation areas on heat maps overlapped with the tumor in 350 of 360 CTs (97%). Performance was high across tumor stages (sensitivity of 0.80, 0.87, 0.95, and 1.0 on T1 through T4 stages, respectively), comparable for hypodense vs isodense tumors (sensitivity: 0.90 vs 0.82), different age, sex, CT slice thicknesses, and vendors (all P > .05), and generalizable on both the simulated cohort (accuracy, 0.95 [95% 0.94-0.95]; AUROC curve, 0.97 [95% CI, 0.94-0.99]) and public data sets (accuracy, 0.86 [95% CI, 0.82-0.90]; AUROC curve, 0.90 [95% CI, 0.86-0.95]). Despite being exclusively trained on diagnostic CTs with larger tumors, the model could detect occult PDA on prediagnostic CTs (accuracy, 0.84 [95% CI, 0.79-0.88]; AUROC curve, 0.91 [95% CI, 0.86-0.94]; sensitivity, 0.75 [95% CI, 0.67-0.84]; and specificity, 0.90 [95% CI, 0.85-0.95]) at a median 475 days (range, 93-1082 days) before clinical diagnosis. CONCLUSIONS This automated artificial intelligence model trained on a large and diverse data set shows high accuracy and generalizable performance for detection of PDA on diagnostic CTs as well as for visually occult PDA on prediagnostic CTs. Prospective validation with blood-based biomarkers is warranted to assess the potential for early detection of sporadic PDA in high-risk individuals.
Collapse
Affiliation(s)
| | - Garima Suman
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | - Cole Cook
- Division of Medical Imaging Technology Services, Mayo Clinic, Rochester, Minnesota
| | - Jason R Klug
- Division of Medical Imaging Technology Services, Mayo Clinic, Rochester, Minnesota
| | - Anurima Patra
- Department of Radiology, Tata Medical Center, Kolkata, India
| | - Hala Khasawneh
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Mark J Truty
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Shounak Majumder
- Department of Gastroenterology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Suresh T Chari
- Department of Gastroenterology, Mayo Clinic, Rochester, Minnesota
| | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
21
|
Wöltjen MM, Kröger JR. [Current CT developments in imaging of pancreatic diseases]. RADIOLOGIE (HEIDELBERG, GERMANY) 2023; 63:878-885. [PMID: 37947865 DOI: 10.1007/s00117-023-01230-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Diseases of the pancreas are often diagnosed late and can have fatal consequences for patients. PURPOSE Current computed tomography (CT) developments in imaging of pancreatic diseases. MATERIALS AND METHODS Evaluation of numerous studies, especially considering modern CT techniques such as dual-energy CT and photon-counting CT but also artificial intelligence (AI) algorithms for disease detection. RESULTS Spectral imaging using dual-energy CT and photon-counting CT offers numerous advantages in the detection of pancreatic disease and can thus improve diagnostic performance but also provide additional information on any therapeutic response. Likewise, advances in the development of AI algorithms are improving diagnostic performance. CONCLUSION In the future, we can expect increasingly improved detection of pancreatic diseases, thereby enabling patients to be treated more quickly, which will consequently result in improved outcomes.
Collapse
|
22
|
Zins M. Invited Commentary: Failures in Imaging Diagnosis of Pancreatic Cancer: Can We Avoid Them and How? Radiographics 2023; 43:e230217. [PMID: 37824410 DOI: 10.1148/rg.230217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Affiliation(s)
- Marc Zins
- From the Department of Medical Imaging, Hôpital Paris Saint-Joseph, 185 Rue Raymond Losserand, 75014 Paris, France
| |
Collapse
|
23
|
Chu LC, Ahmed T, Blanco A, Javed A, Weisberg EM, Kawamoto S, Hruban RH, Kinzler KW, Vogelstein B, Fishman EK. Radiologists' Expectations of Artificial Intelligence in Pancreatic Cancer Imaging: How Good Is Good Enough? J Comput Assist Tomogr 2023; 47:845-849. [PMID: 37948357 PMCID: PMC10823576 DOI: 10.1097/rct.0000000000001503] [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] [Indexed: 07/29/2023]
Abstract
BACKGROUND Existing (artificial intelligence [AI]) tools in radiology are modeled without necessarily considering the expectations and experience of the end user-the radiologist. The literature is scarce on the tangible parameters that AI capabilities need to meet for radiologists to consider them useful tools. OBJECTIVE The purpose of this study is to explore radiologists' attitudes toward AI tools in pancreatic cancer imaging and to quantitatively assess their expectations of these tools. METHODS A link to the survey was posted on the www.ctisus.com website, advertised in the www.ctisus.com email newsletter, and publicized on LinkedIn, Facebook, and Twitter accounts. This survey asked participants about their demographics, practice, and current attitudes toward AI. They were also asked about their expectations of what constitutes a clinically useful AI tool. The survey consisted of 17 questions, which included 9 multiple choice questions, 2 Likert scale questions, 4 binary (yes/no) questions, 1 rank order question, and 1 free text question. RESULTS A total of 161 respondents completed the survey, yielding a response rate of 46.3% of the total 348 clicks on the survey link. The minimum acceptable sensitivity of an AI program for the detection of pancreatic cancer chosen by most respondents was either 90% or 95% at a specificity of 95%. The minimum size of pancreatic cancer that most respondents would find an AI useful at detecting was 5 mm. Respondents preferred AI tools that demonstrated greater sensitivity over those with greater specificity. Over half of respondents anticipated incorporating AI tools into their clinical practice within the next 5 years. CONCLUSION Radiologists are open to the idea of integrating AI-based tools and have high expectations regarding the performance of these tools. Consideration of radiologists' input is important to contextualize expectations and optimize clinical adoption of existing and future AI tools.
Collapse
Affiliation(s)
- Linda C. Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| | - Taha Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| | - Alejandra Blanco
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| | - Ammar Javed
- Department of Surgery, New York University Grossman School of Medicine, New York, NY
| | - Edmund M. Weisberg
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| | - Ralph H. Hruban
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kenneth W. Kinzler
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Bert Vogelstein
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, Maryland
| |
Collapse
|
24
|
Abi Nader C, Vetil R, Wood LK, Rohe MM, Bône A, Karteszi H, Vullierme MP. Automatic Detection of Pancreatic Lesions and Main Pancreatic Duct Dilatation on Portal Venous CT Scans Using Deep Learning. Invest Radiol 2023; 58:791-798. [PMID: 37289274 DOI: 10.1097/rli.0000000000000992] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
OBJECTIVES This study proposes and evaluates a deep learning method to detect pancreatic neoplasms and to identify main pancreatic duct (MPD) dilatation on portal venous computed tomography scans. MATERIALS AND METHODS A total of 2890 portal venous computed tomography scans from 9 institutions were acquired, among which 2185 had a pancreatic neoplasm and 705 were healthy controls. Each scan was reviewed by one in a group of 9 radiologists. Physicians contoured the pancreas, pancreatic lesions if present, and the MPD if visible. They also assessed tumor type and MPD dilatation. Data were split into a training and independent testing set of 2134 and 756 cases, respectively.A method to detect pancreatic lesions and MPD dilatation was built in 3 steps. First, a segmentation network was trained in a 5-fold cross-validation manner. Second, outputs of this network were postprocessed to extract imaging features: a normalized lesion risk, the predicted lesion diameter, and the MPD diameter in the head, body, and tail of the pancreas. Third, 2 logistic regression models were calibrated to predict lesion presence and MPD dilatation, respectively. Performance was assessed on the independent test cohort using receiver operating characteristic analysis. The method was also evaluated on subgroups defined based on lesion types and characteristics. RESULTS The area under the curve of the model detecting lesion presence in a patient was 0.98 (95% confidence interval [CI], 0.97-0.99). A sensitivity of 0.94 (469 of 493; 95% CI, 0.92-0.97) was reported. Similar values were obtained in patients with small (less than 2 cm) and isodense lesions with a sensitivity of 0.94 (115 of 123; 95% CI, 0.87-0.98) and 0.95 (53 of 56, 95% CI, 0.87-1.0), respectively. The model sensitivity was also comparable across lesion types with values of 0.94 (95% CI, 0.91-0.97), 1.0 (95% CI, 0.98-1.0), 0.96 (95% CI, 0.97-1.0) for pancreatic ductal adenocarcinoma, neuroendocrine tumor, and intraductal papillary neoplasm, respectively. Regarding MPD dilatation detection, the model had an area under the curve of 0.97 (95% CI, 0.96-0.98). CONCLUSIONS The proposed approach showed high quantitative performance to identify patients with pancreatic neoplasms and to detect MPD dilatation on an independent test cohort. Performance was robust across subgroups of patients with different lesion characteristics and types. Results confirmed the interest to combine a direct lesion detection approach with secondary features such as the MPD diameter, thus indicating a promising avenue for the detection of pancreatic cancer at early stages.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marie-Pierre Vullierme
- Department of Radiology, Hospital of Annecy-Genevois, Université Paris-Cité, Paris, France
| |
Collapse
|
25
|
Yao L, Zhang Z, Demir U, Keles E, Vendrami C, Agarunov E, Bolan C, Schoots I, Bruno M, Keswani R, Miller F, Gonda T, Yazici C, Tirkes T, Wallace M, Spampinato C, Bagci U. Radiomics Boosts Deep Learning Model for IPMN Classification. MACHINE LEARNING IN MEDICAL IMAGING. MLMI (WORKSHOP) 2023; 14349:134-143. [PMID: 38274402 PMCID: PMC10810260 DOI: 10.1007/978-3-031-45676-3_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Intraductal Papillary Mucinous Neoplasm (IPMN) cysts are pre-malignant pancreas lesions, and they can progress into pancreatic cancer. Therefore, detecting and stratifying their risk level is of ultimate importance for effective treatment planning and disease control. However, this is a highly challenging task because of the diverse and irregular shape, texture, and size of the IPMN cysts as well as the pancreas. In this study, we propose a novel computer-aided diagnosis pipeline for IPMN risk classification from multi-contrast MRI scans. Our proposed analysis framework includes an efficient volumetric self-adapting segmentation strategy for pancreas delineation, followed by a newly designed deep learning-based classification scheme with a radiomics-based predictive approach. We test our proposed decision-fusion model in multi-center data sets of 246 multi-contrast MRI scans and obtain superior performance to the state of the art (SOTA) in this field. Our ablation studies demonstrate the significance of both radiomics and deep learning modules for achieving the new SOTA performance compared to international guidelines and published studies (81.9% vs 61.3% in accuracy). Our findings have important implications for clinical decision-making. In a series of rigorous experiments on multi-center data sets (246 MRI scans from five centers), we achieved unprecedented performance (81.9% accuracy). The code is available upon publication.
Collapse
Affiliation(s)
- Lanhong Yao
- Department of Radiology, Northwestern University, Chicago IL 60611, USA
| | - Zheyuan Zhang
- Department of Radiology, Northwestern University, Chicago IL 60611, USA
| | - Ugur Demir
- Department of Radiology, Northwestern University, Chicago IL 60611, USA
| | - Elif Keles
- Department of Radiology, Northwestern University, Chicago IL 60611, USA
| | - Camila Vendrami
- Department of Radiology, Northwestern University, Chicago IL 60611, USA
| | | | | | - Ivo Schoots
- Erasmus Medical Center, 3015 GD Rotterdam, Netherlands
| | - Marc Bruno
- Erasmus Medical Center, 3015 GD Rotterdam, Netherlands
| | - Rajesh Keswani
- Department of Radiology, Northwestern University, Chicago IL 60611, USA
| | - Frank Miller
- Department of Radiology, Northwestern University, Chicago IL 60611, USA
| | | | - Cemal Yazici
- University of Illinois Chicago, Chicago, IL 60607
| | - Temel Tirkes
- Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202
| | - Michael Wallace
- Sheikh Shakhbout Medical City, 11001, Abu Dhabi, United Arab Emirates
| | | | - Ulas Bagci
- Department of Radiology, Northwestern University, Chicago IL 60611, USA
| |
Collapse
|
26
|
Mervak BM, Fried JG, Wasnik AP. A Review of the Clinical Applications of Artificial Intelligence in Abdominal Imaging. Diagnostics (Basel) 2023; 13:2889. [PMID: 37761253 PMCID: PMC10529018 DOI: 10.3390/diagnostics13182889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/23/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Artificial intelligence (AI) has been a topic of substantial interest for radiologists in recent years. Although many of the first clinical applications were in the neuro, cardiothoracic, and breast imaging subspecialties, the number of investigated and real-world applications of body imaging has been increasing, with more than 30 FDA-approved algorithms now available for applications in the abdomen and pelvis. In this manuscript, we explore some of the fundamentals of artificial intelligence and machine learning, review major functions that AI algorithms may perform, introduce current and potential future applications of AI in abdominal imaging, provide a basic understanding of the pathways by which AI algorithms can receive FDA approval, and explore some of the challenges with the implementation of AI in clinical practice.
Collapse
Affiliation(s)
| | | | - Ashish P. Wasnik
- Department of Radiology, University of Michigan—Michigan Medicine, 1500 E. Medical Center Dr., Ann Arbor, MI 48109, USA; (B.M.M.); (J.G.F.)
| |
Collapse
|
27
|
Yao L, Zhang Z, Keles E, Yazici C, Tirkes T, Bagci U. A review of deep learning and radiomics approaches for pancreatic cancer diagnosis from medical imaging. Curr Opin Gastroenterol 2023; 39:436-447. [PMID: 37523001 PMCID: PMC10403281 DOI: 10.1097/mog.0000000000000966] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
PURPOSE OF REVIEW Early and accurate diagnosis of pancreatic cancer is crucial for improving patient outcomes, and artificial intelligence (AI) algorithms have the potential to play a vital role in computer-aided diagnosis of pancreatic cancer. In this review, we aim to provide the latest and relevant advances in AI, specifically deep learning (DL) and radiomics approaches, for pancreatic cancer diagnosis using cross-sectional imaging examinations such as computed tomography (CT) and magnetic resonance imaging (MRI). RECENT FINDINGS This review highlights the recent developments in DL techniques applied to medical imaging, including convolutional neural networks (CNNs), transformer-based models, and novel deep learning architectures that focus on multitype pancreatic lesions, multiorgan and multitumor segmentation, as well as incorporating auxiliary information. We also discuss advancements in radiomics, such as improved imaging feature extraction, optimized machine learning classifiers and integration with clinical data. Furthermore, we explore implementing AI-based clinical decision support systems for pancreatic cancer diagnosis using medical imaging in practical settings. SUMMARY Deep learning and radiomics with medical imaging have demonstrated strong potential to improve diagnostic accuracy of pancreatic cancer, facilitate personalized treatment planning, and identify prognostic and predictive biomarkers. However, challenges remain in translating research findings into clinical practice. More studies are required focusing on refining these methods, addressing significant limitations, and developing integrative approaches for data analysis to further advance the field of pancreatic cancer diagnosis.
Collapse
Affiliation(s)
- Lanhong Yao
- Machine & Hybrid Intelligence Lab, Department of Radiology, Northwestern University
| | - Zheyuan Zhang
- Machine & Hybrid Intelligence Lab, Department of Radiology, Northwestern University
| | - Elif Keles
- Machine & Hybrid Intelligence Lab, Department of Radiology, Northwestern University
| | - Cemal Yazici
- Division of Gastroentrrology and Hepatology, University of Illinois Chicago, Chicago, Illinois
| | - Temel Tirkes
- Department of Radiology & Imaging Sciences, Medicine and Urology, Indiana University School of Medicine, Indianapolis, Indianapolis, USA
| | - Ulas Bagci
- Machine & Hybrid Intelligence Lab, Department of Radiology, Northwestern University
| |
Collapse
|
28
|
Ahmed TM, Kawamoto S, Hruban RH, Fishman EK, Soyer P, Chu LC. A primer on artificial intelligence in pancreatic imaging. Diagn Interv Imaging 2023; 104:435-447. [PMID: 36967355 DOI: 10.1016/j.diii.2023.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 06/18/2023]
Abstract
Artificial Intelligence (AI) is set to transform medical imaging by leveraging the vast data contained in medical images. Deep learning and radiomics are the two main AI methods currently being applied within radiology. Deep learning uses a layered set of self-correcting algorithms to develop a mathematical model that best fits the data. Radiomics converts imaging data into mineable features such as signal intensity, shape, texture, and higher-order features. Both methods have the potential to improve disease detection, characterization, and prognostication. This article reviews the current status of artificial intelligence in pancreatic imaging and critically appraises the quality of existing evidence using the radiomics quality score.
Collapse
Affiliation(s)
- Taha M Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ralph H Hruban
- Sol Goldman Pancreatic Research Center, Department of Pathology, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Philippe Soyer
- Université Paris Cité, Faculté de Médecine, Department of Radiology, Hôpital Cochin-APHP, 75014, 75006, Paris, France, 7501475006
| | - Linda C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
29
|
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: 4.0] [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
|
30
|
Karar ME, El-Fishawy N, Radad M. Automated classification of urine biomarkers to diagnose pancreatic cancer using 1-D convolutional neural networks. J Biol Eng 2023; 17:28. [PMID: 37069681 PMCID: PMC10111836 DOI: 10.1186/s13036-023-00340-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/13/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Early diagnosis of Pancreatic Ductal Adenocarcinoma (PDAC) is the main key to surviving cancer patients. Urine proteomic biomarkers which are creatinine, LYVE1, REG1B, and TFF1 present a promising non-invasive and inexpensive diagnostic method of the PDAC. Recent utilization of both microfluidics technology and artificial intelligence techniques enables accurate detection and analysis of these biomarkers. This paper proposes a new deep-learning model to identify urine biomarkers for the automated diagnosis of pancreatic cancers. The proposed model is composed of one-dimensional convolutional neural networks (1D-CNNs) and long short-term memory (LSTM). It can categorize patients into healthy pancreas, benign hepatobiliary disease, and PDAC cases automatically. RESULTS Experiments and evaluations have been successfully done on a public dataset of 590 urine samples of three classes, which are 183 healthy pancreas samples, 208 benign hepatobiliary disease samples, and 199 PDAC samples. The results demonstrated that our proposed 1-D CNN + LSTM model achieved the best accuracy score of 97% and the area under curve (AUC) of 98% versus the state-of-the-art models to diagnose pancreatic cancers using urine biomarkers. CONCLUSION A new efficient 1D CNN-LSTM model has been successfully developed for early PDAC diagnosis using four proteomic urine biomarkers of creatinine, LYVE1, REG1B, and TFF1. This developed model showed superior performance on other machine learning classifiers in previous studies. The main prospect of this study is the laboratory realization of our proposed deep classifier on urinary biomarker panels for assisting diagnostic procedures of pancreatic cancer patients.
Collapse
Affiliation(s)
- Mohamed Esmail Karar
- Department of Industrial Electronics and Control Engineering, Faculty of Electronic Engineering, Menoufia University, Al Minufiyah, Egypt
| | - Nawal El-Fishawy
- Department of Computer Science and Engineering, Faculty of Electronic Engineering, Menoufia University, Al Minufiyah, Egypt
| | - Marwa Radad
- Department of Computer Science and Engineering, Faculty of Electronic Engineering, Menoufia University, Al Minufiyah, Egypt.
| |
Collapse
|
31
|
Fowler KJ. Gastrointestinal Imaging: What Has Shaped and What Will Shape Our Field Going Forward. Radiology 2023; 307:e223251. [PMID: 36916893 DOI: 10.1148/radiol.223251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Affiliation(s)
- Kathryn J Fowler
- From the Department of Diagnostic Radiology, Division of Body Imaging, University of California-San Diego, 6206 Lakewood St, San Diego, CA 92122
| |
Collapse
|
32
|
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: 3] [Impact Index Per Article: 3.0] [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
|
33
|
Scherübl H. [Prevention of pancreatic cancer]. Dtsch Med Wochenschr 2023; 148:246-252. [PMID: 36848888 DOI: 10.1055/a-1975-2366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
The incidence of pancreatic cancer is rising in Germany. At present pancreatic cancer is the third commonest cause of cancer death but is expected to become the second in 2030 and finally the leading cause of cancer death in 2050. Pancreatic ductal adenocarcinoma (PC) is generally diagnosed at far advanced stages and 5-year-survival has remained poor. Modifiable risk factors of PC are tobacco smoking, excess body weight, alcohol use, type 2-diabetes and the metabolic syndrome. Smoking cessation and -in case of obesity- intentional weight loss can reduce PC risk by as much as 50 %. Early detection of asymptomatic sporadic PC at stage IA - stage IA-PC now has a 5-year-survival rate of about 80 %- has become a realistic chance for people older than 50 years with new-onset diabetes.
Collapse
Affiliation(s)
- Hans Scherübl
- Klinik für Gastroenterologie, Gastrointestinale Onkologie und Infektiologie, Vivantes Klinikum am Urban, 10967 Berlin
| |
Collapse
|
34
|
Moy L. Top Publications in Radiology, 2022. Radiology 2023; 306:e222914. [PMID: 36625749 DOI: 10.1148/radiol.222914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
35
|
Mazer BL, Lee JW, Roberts NJ, Chu LC, Lennon AM, Klein AP, Eshleman JR, Fishman EK, Canto MI, Goggins MG, Hruban RH. Screening for pancreatic cancer has the potential to save lives, but is it practical? Expert Rev Gastroenterol Hepatol 2023; 17:555-574. [PMID: 37212770 PMCID: PMC10424088 DOI: 10.1080/17474124.2023.2217354] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/21/2023] [Accepted: 05/19/2023] [Indexed: 05/23/2023]
Abstract
INTRODUCTION Most patients with pancreatic cancer present with advanced stage, incurable disease. However, patients with high-grade precancerous lesions and many patients with low-stage disease can be cured with surgery, suggesting that early detection has the potential to improve survival. While serum CA19.9 has been a long-standing biomarker used for pancreatic cancer disease monitoring, its low sensitivity and poor specificity have driven investigators to hunt for better diagnostic markers. AREAS COVERED This review will cover recent advances in genetics, proteomics, imaging, and artificial intelligence, which offer opportunities for the early detection of curable pancreatic neoplasms. EXPERT OPINION From exosomes, to circulating tumor DNA, to subtle changes on imaging, we know much more now about the biology and clinical manifestations of early pancreatic neoplasia than we did just five years ago. The overriding challenge, however, remains the development of a practical approach to screen for a relatively rare, but deadly, disease that is often treated with complex surgery. It is our hope that future advances will bring us closer to an effective and financially sound approach for the early detection of pancreatic cancer and its precursors.
Collapse
Affiliation(s)
- Benjamin L. Mazer
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jae W. Lee
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nicholas J. Roberts
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Linda C. Chu
- Department of Radiology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anne Marie Lennon
- Department of Medicine, Division of Gastroenterology and Hepatology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alison P. Klein
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James R. Eshleman
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K. Fishman
- Department of Radiology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcia Irene Canto
- Department of Medicine, Division of Gastroenterology and Hepatology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael G. Goggins
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ralph H. Hruban
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
36
|
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: 6] [Impact Index Per Article: 3.0] [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
|