1
|
Gadour E, Miutescu B, Hassan Z, Aljahdli ES, Raees K. Advancements in the diagnosis of biliopancreatic diseases: A comparative review and study on future insights. World J Gastrointest Endosc 2025; 17:103391. [DOI: 10.4253/wjge.v17.i4.103391] [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: 11/26/2024] [Revised: 02/19/2025] [Accepted: 03/08/2025] [Indexed: 04/14/2025] Open
Abstract
Owing to the complex and often asymptomatic presentations, the diagnosis of biliopancreatic diseases, including pancreatic and biliary malignancies, remains challenging. Recent technological advancements have remarkably improved the diagnostic accuracy and patient outcomes in these diseases. This review explores key advancements in diagnostic modalities, including biomarkers, imaging techniques, and artificial intelligence (AI)-based technologies. Biomarkers, such as cancer antigen 19-9, KRAS mutations, and inflammatory markers, provide crucial insights into disease progression and treatment responses. Advanced imaging modalities include enhanced computed tomography (CT), positron emission tomography-CT, magnetic resonance cholangiopancreatography, and endoscopic ultrasound. AI integration in imaging and pathology has enhanced diagnostic precision through deep learning algorithms that analyze medical images, automate routine diagnostic tasks, and provide predictive analytics for personalized treatment strategies. The applications of these technologies are diverse, ranging from early cancer detection to therapeutic guidance and real-time imaging. Biomarker-based liquid biopsies and AI-assisted imaging tools are essential for non-invasive diagnostics and individualized patient management. Furthermore, AI-driven models are transforming disease stratification, thus enhancing risk assessment and decision-making. Future studies should explore standardizing biomarker validation, improving AI-driven diagnostics, and expanding the accessibility of advanced imaging technologies in resource-limited settings. The continued development of non-invasive diagnostic techniques and precision medicine approaches is crucial for optimizing the detection and management of biliopancreatic diseases. Collaborative efforts between clinicians, researchers, and industry stakeholders will be pivotal in applying these advancements in clinical practice.
Collapse
Affiliation(s)
- Eyad Gadour
- Multiorgan Transplant Centre of Excellence, Liver Transplantation Unit, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia
- Internal Medicine, Zamzam University College, School of Medicine, Khartoum 11113, Sudan
| | - Bogdan Miutescu
- Department of Gastroenterology and Hepatology, Victor Babes University of Medicine and Pharmacy, Timisoara 300041, Romania
- Advanced Regional Research Center in Gastroenterology and Hepatology, Victor Babes University of Medicine and Pharmacy, Timisoara 30041, Romania
| | - Zeinab Hassan
- Department of Internal Medicine, Stockport Hospitals NHS Foundation Trust, Manchester SK2 7JE, United Kingdom
| | - Emad S Aljahdli
- Gastroenterology Division, King Abdulaziz University, Faculty of Medicine, Jeddah 21589, Saudi Arabia
- Gastrointestinal Oncology Unit, King Abdulaziz University Hospital, Jeddah 22252, Saudi Arabia
| | - Khurram Raees
- Department of Gastroenterology and Hepatology, Royal Blackburn Hospital, Blackburn BB2 3HH, United Kingdom
| |
Collapse
|
2
|
Peduzzi G, Felici A, Pellungrini R, Campa D. Explainable machine learning identifies a polygenic risk score as a key predictor of pancreatic cancer risk in the UK Biobank. Dig Liver Dis 2025; 57:915-922. [PMID: 39632152 DOI: 10.1016/j.dld.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Predicting the risk of developing pancreatic ductal adenocarcinoma (PDAC) is of paramount importance, given its high mortality rate. Current PDAC risk prediction models rely on a limited number of variables, do not include genetics, and have a modest accuracy. AIM This study aimed to develop an interpretable PDAC risk prediction model, based on machine learning (ML). METHODS Five ML models (Adaptive Boosting, eXtreme Gradient Boosting, CatBoost, Deep Forest and Random Forest) built on 56 exposome variables and a polygenic risk score (PRS) were tested in 654 PDAC cases and 1,308 controls of the UK Biobank. Additionally, SHapley Additive exPlanation (SHAP) and Global model Interpretation via the Recursive Partitioning (Girp) were employed to explain the models. RESULTS All models provided similar performance, but based on recall the best was CatBoost (77.10 %). SHAP highlighted age and the PRS as primary contributors across all models. Girp developed rules to discern cases from controls, identifying age, PRS, and pancreatitis in most of the rules. CONCLUSION The predictive models tested have exhibited good performance, indicating their potential application in the clinical field in the near future, with the PRS playing a key role in identifying high-risk individuals as demonstrated by the explainers.
Collapse
Affiliation(s)
- Giulia Peduzzi
- Department of Biology, University of Pisa, Via Luca Ghini, 13 - 56126, Pisa, Italy.
| | - Alessio Felici
- Department of Biology, University of Pisa, Via Luca Ghini, 13 - 56126, Pisa, Italy.
| | - Roberto Pellungrini
- Classe di scienze, Scuola Normale Superiore, Piazza dei Cavalieri, 7 - 56126, Pisa, Italy.
| | - Daniele Campa
- Department of Biology, University of Pisa, Via Luca Ghini, 13 - 56126, Pisa, Italy.
| |
Collapse
|
3
|
Abdul Rasool Hassan B, Mohammed AH, Hallit S, Malaeb D, Hosseini H. Exploring the role of artificial intelligence in chemotherapy development, cancer diagnosis, and treatment: present achievements and future outlook. Front Oncol 2025; 15:1475893. [PMID: 39990683 PMCID: PMC11843581 DOI: 10.3389/fonc.2025.1475893] [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: 08/04/2024] [Accepted: 01/13/2025] [Indexed: 02/25/2025] Open
Abstract
Background Artificial intelligence (AI) has emerged as a transformative tool in oncology, offering promising applications in chemotherapy development, cancer diagnosis, and predicting chemotherapy response. Despite its potential, debates persist regarding the predictive accuracy of AI technologies, particularly machine learning (ML) and deep learning (DL). Objective This review aims to explore the role of AI in forecasting outcomes related to chemotherapy development, cancer diagnosis, and treatment response, synthesizing current advancements and identifying critical gaps in the field. Methods A comprehensive literature search was conducted across PubMed, Embase, Web of Science, and Cochrane databases up to 2023. Keywords included "Artificial Intelligence (AI)," "Machine Learning (ML)," and "Deep Learning (DL)" combined with "chemotherapy development," "cancer diagnosis," and "cancer treatment." Articles published within the last four years and written in English were included. The Prediction Model Risk of Bias Assessment tool was utilized to assess the risk of bias in the selected studies. Conclusion This review underscores the substantial impact of AI, including ML and DL, on cancer diagnosis, chemotherapy innovation, and treatment response for both solid and hematological tumors. Evidence from recent studies highlights AI's potential to reduce cancer-related mortality by optimizing diagnostic accuracy, personalizing treatment plans, and improving therapeutic outcomes. Future research should focus on addressing challenges in clinical implementation, ethical considerations, and scalability to enhance AI's integration into oncology care.
Collapse
Affiliation(s)
| | | | - Souheil Hallit
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik, Jounieh, Lebanon
- Department of Psychology, College of Humanities, Effat University, Jeddah, Saudi Arabia
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | - Diana Malaeb
- College of Pharmacy, Gulf Medical University, Ajman, United Arab Emirates
| | - Hassan Hosseini
- Institut Coeur et Cerveau de l’Est Parisien (ICCE), UPEC-University Paris-Est, Creteil, France
- RAMSAY SANTÉ, Hôpital Paul D’Egine (HPPE), Champigny sur Marne, France
| |
Collapse
|
4
|
Zhou H, Zheng Z, Fan C, Zhou Z. Mechanisms and strategies of immunosenescence effects on non-small cell lung cancer (NSCLC) treatment: A comprehensive analysis and future directions. Semin Cancer Biol 2025; 109:44-66. [PMID: 39793777 DOI: 10.1016/j.semcancer.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Non-small cell lung cancer (NSCLC), the most prevalent form of lung cancer, remains a leading cause of cancer-related mortality worldwide, particularly among elderly individuals. The phenomenon of immunosenescence, characterized by the progressive decline in immune cell functionality with aging, plays a pivotal role in NSCLC progression and contributes to the diminished efficacy of therapeutic interventions in older patients. Immunosenescence manifests through impaired immune surveillance, reduced cytotoxic responses, and increased chronic inflammation, collectively fostering a pro-tumorigenic microenvironment. This review provides a comprehensive analysis of the molecular, cellular, and genetic mechanisms of immunosenescence and its impact on immune surveillance and the tumor microenvironment (TME) in NSCLC. We explore how aging affects various immune cells, including T cells, B cells, NK cells, and macrophages, and how these changes compromise the immune system's ability to detect and eliminate tumor cells. Furthermore, we address the challenges posed by immunosenescence to current therapeutic strategies, particularly immunotherapy, which faces significant hurdles in elderly patients due to immune dysfunction. The review highlights emerging technologies, such as single-cell sequencing and CRISPR-Cas9, which offer new insights into immunosenescence and its potential as a therapeutic target. Finally, we outline future research directions, including strategies for rejuvenating the aging immune system and optimizing immunotherapy for older NSCLC patients, with the goal of improving treatment efficacy and survival outcomes. These efforts hold promise for the development of more effective, personalized therapies for elderly patients with NSCLC.
Collapse
Affiliation(s)
- Huatao Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Zilong Zheng
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| | - Zijing Zhou
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| |
Collapse
|
5
|
Nishat SMH, Shahid Tanweer A, Alshamsi B, Shaheen MH, Shahid Tanveer A, Nishat A, Alharbat Y, Alaboud A, Almazrouei M, Ali-Mohamed RA. Artificial Intelligence: A New Frontier in Rare Disease Early Diagnosis. Cureus 2025; 17:e79487. [PMID: 40135033 PMCID: PMC11933855 DOI: 10.7759/cureus.79487] [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] [Accepted: 02/22/2025] [Indexed: 03/27/2025] Open
Abstract
Rare diseases present significant challenges, including delays in diagnosis, inadequate treatment responses, and difficulties in monitoring. These challenges arise from the complexity of symptoms, limited medical expertise, and insufficient diagnostic tools. Artificial Intelligence (AI) has gained attention for its potential to improve healthcare, particularly in diagnosing complex conditions. By analyzing large datasets, recognizing patterns, and integrating clinical information, AI can refine diagnostic accuracy, enhance treatment strategies, and improve patient outcomes. This literature review examines AI applications in three key areas of rare disease diagnosis: genetic analysis, imaging-based phenotyping, and natural language processing (NLP) for clinical data extraction. AI-driven advancements in these domains have been explored to improve disease detection and management. However, several challenges persist, including limited data availability, algorithm transparency, privacy considerations, and ethical concerns. Efforts such as data augmentation and transfer learning are being explored to address these issues and expand AI's role in clinical practice. By refining diagnostic processes and optimizing treatment strategies, AI has the potential to improve the management of rare diseases. This review critically examines AI's role in rare disease diagnosis, with a particular emphasis on its applications in genetic analysis, imaging-based phenotyping, and NLP, while also addressing key challenges and future directions for clinical integration.
Collapse
Affiliation(s)
| | - Ammar Shahid Tanweer
- Internal Medicine, RAK Medical & Health Sciences University, Ras Al Khaimah, ARE
| | - Bashayer Alshamsi
- Medicine, RAK Medical & Health Sciences University, Ras Al Khaimah, ARE
| | - Majd H Shaheen
- Medicine, RAK Medical & Health Sciences University, Ras Al Khaimah, ARE
| | | | - Aroob Nishat
- Internal Medicine, RAK Medical & Health Sciences University, Ras Al Khaimah, ARE
| | - Yaman Alharbat
- Internal Medicine, RAK Medical & Health Sciences University, Ras Al Khaimah, ARE
| | - Ahmad Alaboud
- Internal Medicine, RAK Medical & Health Sciences University, Ras Al Khaimah, ARE
| | - Mahra Almazrouei
- Medicine, RAK Medical & Health Sciences University, Ras Al Khaimah, ARE
| | - Raghad A Ali-Mohamed
- Internal Medicine, RAK Medical & Health Sciences University, Ras Al Khaimah, ARE
| |
Collapse
|
6
|
Xiong L, Li D, Xiao G, Tan S, Xu L, Wang G. HSP70 Promotes Pancreatic Cancer Cell Epithelial-Mesenchymal Transformation and Growth Via the NF-κB Signaling Pathway. Pancreas 2025; 54:e89-e96. [PMID: 39352012 DOI: 10.1097/mpa.0000000000002398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
OBJECTIVE To study the effects of HSP70 on proliferation, migration, invasion, and epithelial-mesenchymal transformation (EMT) of pancreatic cancer cells and explore its underlying mechanisms. METHODS Pancreatic cancer cell models with reduced HSP70 or increased HSP70 expression were established. Reverse transcription quantitative polymerase chain reaction and Western blot assays were used to determine mRNA and protein levels of HSP70, IKK/IκBa/NF-κB signaling pathway-related genes, and EMT markers. CCK-8 and cell cloning assays were used to evaluate cell proliferation and cloning abilities. Transwell and wound healing assays were used to assess the invasive and migratory properties of cells. Electrophoresis mobility shift assay (EMSA) and luciferase reporter assays were conducted to analyze NF-κB's promoter binding and transcriptional activities. RESULTS HSP70 knockdown inhibited p-p65 nuclear translocation, the expression of p-p65, p-IKKα/β, p-IκBα, N-cadherin, Vimentin and Twist, NF-κB's promoter binding and transcriptional activities, pancreatic cancer cell proliferation, cloning, migration and invasion, while increased E-cadherin levels. HSP70 overexpression took the opposite effects. NF-κB signaling pathway modulation reversed EMT changes induced by altered HSP70 expression levels. rhHSP70 increased p-IKKα/β and p-IκBα protein levels. CONCLUSIONS HSP70 promotes EMT and enhances pancreatic cancer cell proliferation, migration, and invasion by activating NF-κB pathway.
Collapse
Affiliation(s)
- Liumei Xiong
- From the Department of Gastroenterology, Pingxing Hospital, Southern Medical University, Pingxiang, China
| | - Danming Li
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gui Xiao
- Department of International School of Nursing, Hainan Medical University, Haikou, China
| | - Sipin Tan
- Sepsis Translational Medicine, Key Lab of Hunan, Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Linfang Xu
- From the Department of Gastroenterology, Pingxing Hospital, Southern Medical University, Pingxiang, China
| | | |
Collapse
|
7
|
Tan Z, Meng Y, Wu Y, Zhen J, He H, Pu Y, Zhang J, Dong W. The burden and temporal trend of early onset pancreatic cancer based on the GBD 2021. NPJ Precis Oncol 2025; 9:32. [PMID: 39880919 PMCID: PMC11779834 DOI: 10.1038/s41698-025-00820-0] [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: 10/03/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
In the context of the global increase in early-onset tumours, investigating the global disease burden caused by early-onset pancreatic cancer (EOPC) is imperative. Data on the burden of EOPC were obtained from the Global Burden of Disease Study 2021. A joinpoint regression model was used to analyse the temporal trend of the EOPC burden, and an age‒period‒cohort (APC) model was used to analyse the influence of age, period, and birth cohort on burden trends. Globally, the number of EOPC cases increased from 24,480 to 42,254, and the number of deaths increased from 17,193 to 26,996 between 1990 and 2021. The results of the APC model showed that the burden of EOPC increases with increasing age, whereas the variations in period and cohort effects exhibited a complex pattern across different sociodemographic index regions. Consequently, the disease burden of EOPC is increasing worldwide, highlighting the need for effective interventions.
Collapse
Affiliation(s)
- Zongbiao Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yang Meng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yanrui Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Junhai Zhen
- Department of General Practice, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, China
| | - Haodong He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yu Pu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Jixiang Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| |
Collapse
|
8
|
Nadeem A, Ashraf R, Mahmood T, Parveen S. Automated CAD system for early detection and classification of pancreatic cancer using deep learning model. PLoS One 2025; 20:e0307900. [PMID: 39752442 PMCID: PMC11698441 DOI: 10.1371/journal.pone.0307900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/10/2024] [Indexed: 01/06/2025] Open
Abstract
Accurate diagnosis of pancreatic cancer using CT scan images is critical for early detection and treatment, potentially saving numerous lives globally. Manual identification of pancreatic tumors by radiologists is challenging and time-consuming due to the complex nature of CT scan images and variations in tumor shape, size, and location of the pancreatic tumor also make it challenging to detect and classify different types of tumors. Thus, to address this challenge we proposed a four-stage framework of computer-aided diagnosis systems. In the preprocessing stage, the input image resizes into 227 × 227 dimensions then converts the RGB image into a grayscale image, and enhances the image by removing noise without blurring edges by applying anisotropic diffusion filtering. In the segmentation stage, the preprocessed grayscale image a binary image is created based on a threshold, highlighting the edges by Sobel filtering, and watershed segmentation to segment the tumor region and we also implement the U-Net method for segmentation. Then refine the geometric structure of the image using morphological operation and extracting the texture features from the image using a gray-level co-occurrence matrix computed by analyzing the spatial relationship of pixel intensities in the refined image, counting the occurrences of pixel pairs with specific intensity values and spatial relationships. The detection stage analyzes the tumor region's extracted features characteristics by labeling the connected components and selecting the region with the highest density to locate the tumor area, achieving a good accuracy of 99.64%. In the classification stage, the system classifies the detected tumor into the normal, pancreatic tumor, then into benign, pre-malignant, or malignant using a proposed reduced 11-layer AlexNet model. The classification stage attained an accuracy level of 98.72%, an AUC of 0.9979, and an overall system average processing time of 1.51 seconds, demonstrating the capability of the system to effectively and efficiently identify and classify pancreatic cancers.
Collapse
Affiliation(s)
- Abubakar Nadeem
- Department of Computer Science, National Textile University, Faisalabad, Pakistan
| | - Rahan Ashraf
- Department of Computer Science, National Textile University, Faisalabad, Pakistan
| | - Toqeer Mahmood
- Department of Computer Science, National Textile University, Faisalabad, Pakistan
| | - Sajida Parveen
- Department of Computer Science, National Textile University, Faisalabad, Pakistan
| |
Collapse
|
9
|
Corallo C, Al-Adhami AS, Jamieson N, Valle J, Radhakrishna G, Moir J, Albazaz R. An update on pancreatic cancer imaging, staging, and use of the PACT-UK radiology template pre- and post-neoadjuvant treatment. Br J Radiol 2025; 98:13-26. [PMID: 39460945 DOI: 10.1093/bjr/tqae217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/01/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma continues to have a poor prognosis, although recent advances in neoadjuvant treatments (NATs) have provided some hope. Imaging assessment of suspected tumours can be challenging and requires a specific approach, with pancreas protocol CT being the primary imaging modality for staging with other modalities used as problem-solving tools to facilitate appropriate management. Imaging assessment post NAT can be particularly difficult due to a current lack of robust radiological criteria to predict response and differentiate treatment induced fibrosis/inflammation from residual tumour. This review aims to provide an update of pancreatic ductal adenocarcinoma with particular focus on three points: tumour staging pre- and post-NAT including vascular assessment, structured reporting with introduction of the PAncreatic Cancer reporting Template-UK (PACT-UK) radiology template, and the potential future role of artificial intelligence in the diagnosis and staging of pancreatic cancer.
Collapse
Affiliation(s)
- Carmelo Corallo
- Department of Radiology, St James's University Hospital, Leeds LS9 7TF, United Kingdom
| | - Abdullah S Al-Adhami
- Department of Radiology, Glasgow Royal Infirmary, Glasgow G31 2ER, United Kingdom
| | - Nigel Jamieson
- HPB Unit, Glasgow Royal Infirmary, Glasgow G31 2ER, United Kingdom
| | - Juan Valle
- Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, United Kingdom
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4 BX, United Kingdom
| | | | - John Moir
- HPB Unit, Freeman Hospital, Newcastle Upon Tyne NE7 7DN, United Kingdom
| | - Raneem Albazaz
- Department of Radiology, St James's University Hospital, Leeds LS9 7TF, United Kingdom
- University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
10
|
Li Z, Wang C, Li J, Wang X, Li X, Yu T, Zhou J, Wang X, Zeng M, Sun H. Identification of SMAD4-mutated pancreatic ductal adenocarcinoma using preoperative contrast-enhanced MRI and clinical characteristics. BMC Med Imaging 2024; 24:349. [PMID: 39716095 DOI: 10.1186/s12880-024-01539-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024] Open
Abstract
AIM To assess the value of preoperatively contrast-enhanced MRI and clinical characteristics for identification of SMAD4-mutated pancreatic ductal adenocarcinoma (PDAC) patients. MATERIALS AND METHODS This retrospective study included patients with surgically confirmed PDAC from January 2016 to December 2022. Based on immunostaining results indicating the mutation of SMAD4, the enrolled participants were grouped into SMAD4-mutated PDAC and non-SMAD4-mutated PDAC. Contrast-enhanced MRI findings, clinical-pathological characteristics, and prognosis were recorded and reviewed. The pathological findings and clinical prognosis were compared between the two groups. Uni- and multivariable logistic regression analyses were further performed to determine the radiological and clinical predictive factors for the mutation of SMAD4. RESULTS In total, 428 PDAC patients were enrolled and analyzed, who were grouped as SMAD4-mutated PDAC (n = 224) and non-SMAD4-mutated PDAC (n = 204). SMAD4-mutated PDAC demonstrated higher frequency of pathological fatty infiltration (83.4% vs. 74.2%, P = 0.016), peripheral nerve infiltration (84.4% vs. 76.5%, P = 0.039). and higher recurrence rates (43.6% vs. 58.9%, P = 0.045) than non-SMAD4-mutated PDAC. The 3-year recurrence-free survival rates were worse for SMAD4-mutated PDAC (28.7% vs. 39.1%). In multivariable logistic regression analyses, CA19-9 > 100 U/mL (odds ratio [OR] = 1.519, P = 0.041), CBD dilation (OR = 1.564, P = 0.036), and rim enhancement (OR = 1.631, P = 0.025) were independent predictive factors. CONCLUSION Rim enhancement, CBD dilation on contrast-enhanced MRI and higher CA19-9 level are promising radiological and clinical factors for identifying SMAD4-mutated PDAC.
Collapse
Affiliation(s)
- Zhina Li
- Department of Radiology, Zhongshan Hospital, Shanghai Institute of Medical Imaging, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
- Department of Radiology, Linyi Centra Hospital, No.17 Jiankang Road, Linyi City, Shandong Province, 276400, China
| | - Cheng Wang
- Department of Radiology, Zhongshan Hospital, Shanghai Institute of Medical Imaging, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Jianbo Li
- Department of Radiology, Zhongshan Hospital, Shanghai Institute of Medical Imaging, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Xingxing Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Xiang Li
- Department of Pathology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Tianzhu Yu
- Department of Interventional Radiology, Zhongshan Hospital, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China.
| | - Jianjun Zhou
- Department of Radiology, Zhongshan Hospital (Xiamen), Xiamen Municipal Clinical Research Center for Medical Imaging, Fujian Province Key Clinical Specialty for Medical Imaging, Xiamen Key Laboratory of Clinical Transformation of Imaging Big Data and Artificial Intelligence, Fudan University, Xiamen, 361015, China
| | - Xiaolin Wang
- Department of Interventional Radiology, Zhongshan Hospital, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Shanghai Institute of Medical Imaging, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Haitao Sun
- Department of Radiology, Zhongshan Hospital, Shanghai Institute of Medical Imaging, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
11
|
Othman D, Kaleem A. The Intraoperative Role of Artificial Intelligence Within General Surgery: A Systematic Review. Cureus 2024; 16:e73006. [PMID: 39634963 PMCID: PMC11617030 DOI: 10.7759/cureus.73006] [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] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
The role of artificial intelligence has been explored in many industries across the world. The medical field is no exception with studies regarding its use for development of algorithms in cancer screening and its diagnostic utility in clinical radiology. This study aims to review current literature on intraoperative use of artificial intelligence within general surgery to identify the latest developments, the major challenges and the trajectory of this field. A literature search was done on PubMed on May 28, 2024, using the terms: ((artificial intelligence) AND (general surgery)). Only publications in English and studies involving human subjects were considered. Exclusion criteria included duplicate papers, irrelevant titles, abstracts, themes, and non-English papers. A literature search on PubMed yielded 13 relevant articles. Among these, five articles focused on intraoperative guidance, four addressed surgical education and training, and four were survey-based exploring perceptions regarding artificial intelligence. Key themes included the development of artificial intelligence-based autonomous actions during surgery and its role in enhancing surgical training. Limitations identified included restricted data availability, ethical concerns, and a lack of validation tools, which pose significant obstacles to progress in this area. Despite existing limitations, the potential for integrating artificial intelligence into general surgery is promising. Careful attention is needed to overcome challenges and maximize its benefits.
Collapse
Affiliation(s)
- Deema Othman
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, ARE
| | - Ahmad Kaleem
- General Surgery, Mediclinic Parkview Hospital, Dubai, ARE
| |
Collapse
|
12
|
Udriștoiu AL, Podină N, Ungureanu BS, Constantin A, Georgescu CV, Bejinariu N, Pirici D, Burtea DE, Gruionu L, Udriștoiu S, Săftoiu A. Deep learning segmentation architectures for automatic detection of pancreatic ductal adenocarcinoma in EUS-guided fine-needle biopsy samples based on whole-slide imaging. Endosc Ultrasound 2024; 13:335-344. [PMID: 39802107 PMCID: PMC11723688 DOI: 10.1097/eus.0000000000000094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 10/27/2024] [Indexed: 01/16/2025] Open
Abstract
Background EUS-guided fine-needle biopsy is the procedure of choice for the diagnosis of pancreatic ductal adenocarcinoma (PDAC). Nevertheless, the samples obtained are small and require expertise in pathology, whereas the diagnosis is difficult in view of the scarcity of malignant cells and the important desmoplastic reaction of these tumors. With the help of artificial intelligence, the deep learning architectures produce a fast, accurate, and automated approach for PDAC image segmentation based on whole-slide imaging. Given the effectiveness of U-Net in semantic segmentation, numerous variants and improvements have emerged, specifically for whole-slide imaging segmentation. Methods In this study, a comparison of 7 U-Net architecture variants was performed on 2 different datasets of EUS-guided fine-needle biopsy samples from 2 medical centers (31 and 33 whole-slide images, respectively) with different parameters and acquisition tools. The U-Net architecture variants evaluated included some that had not been previously explored for PDAC whole-slide image segmentation. The evaluation of their performance involved calculating accuracy through the mean Dice coefficient and mean intersection over union (IoU). Results The highest segmentation accuracies were obtained using Inception U-Net architecture for both datasets. PDAC tissue was segmented with the overall average Dice coefficient of 97.82% and IoU of 0.87 for Dataset 1, respectively, overall average Dice coefficient of 95.70%, and IoU of 0.79 for Dataset 2. Also, we considered the external testing of the trained segmentation models by performing the cross evaluations between the 2 datasets. The Inception U-Net model trained on Train Dataset 1 performed with the overall average Dice coefficient of 93.12% and IoU of 0.74 on Test Dataset 2. The Inception U-Net model trained on Train Dataset 2 performed with the overall average Dice coefficient of 92.09% and IoU of 0.81 on Test Dataset 1. Conclusions The findings of this study demonstrated the feasibility of utilizing artificial intelligence for assessing PDAC segmentation in whole-slide imaging, supported by promising scores.
Collapse
Affiliation(s)
| | - Nicoleta Podină
- Department of Gastroenterology, Ponderas Academic Hospital, Bucharest, Romania
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Bogdan Silviu Ungureanu
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
- Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Alina Constantin
- Department of Gastroenterology, Ponderas Academic Hospital, Bucharest, Romania
| | | | - Nona Bejinariu
- REGINA MARIA Regional Laboratory, Pathological Anatomy Division, Cluj-Napoca, Romania
| | - Daniel Pirici
- Department of Histology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Daniela Elena Burtea
- Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Lucian Gruionu
- Faculty of Mechanics, University of Craiova, Craiova, Romania
| | - Stefan Udriștoiu
- Faculty of Automation, Computers and Electronics, University of Craiova, Craiova, Romania
| | - Adrian Săftoiu
- Department of Gastroenterology, Ponderas Academic Hospital, Bucharest, Romania
- Department of Gastroenterology and Hepatology, Elias University Emergency Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
13
|
Hu K, Bian C, Yu J, Jiang D, Chen Z, Zhao F, Li H. Construction of a combined prognostic model for pancreatic ductal adenocarcinoma based on deep learning and digital pathology images. BMC Gastroenterol 2024; 24:387. [PMID: 39482576 PMCID: PMC11528996 DOI: 10.1186/s12876-024-03469-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Deep learning has made significant advancements in the field of digital pathology, and the integration of multiple models has further improved accuracy. In this study, we aimed to construct a combined prognostic model using deep learning-extracted features from digital pathology images of pancreatic ductal adenocarcinoma (PDAC) alongside clinical predictive indicators and to explore its prognostic value. METHODS A retrospective analysis was conducted on 142 postoperative pathologically confirmed PDAC cases. These cases were divided into training (n = 114) and testing sets (n = 28) at an 8:2 ratio. Tumor whole-slide imaging features were extracted and screened to construct a pathological risk model based on a pre-trained deep learning model. Clinical and pathological data from the training set were used to select independent predictive factors for PDAC and establish a clinical risk model using LASSO, univariate, and multivariate Cox regression analyses. Based on the pathological and clinical risk models, a combined model was developed. The Harrell concordance index (C-index) was computed to assess the predictive performance of each model for PDAC survival prognosis. RESULTS For the training and testing sets, the C-index values for the clinical risk model were 0.76 and 0.75, respectively; for the pathological risk model, they were 0.82 and 0.73, respectively; and for the combined model, they were 0.86 and 0.77, respectively. The combined model exhibited appropriate calibration at 1-, 3-, and 5-year time points, as well as a superior area under the curve of the receiver operating characteristic curve and clinical net benefit compared to the single models. CONCLUSIONS Integrating the pathological and clinical risk models may provide a higher predictive value for survival prognosis.
Collapse
Affiliation(s)
- Kaixin Hu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Chenyang Bian
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jiayin Yu
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Dawei Jiang
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhangjun Chen
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Fengqing Zhao
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Huangbao Li
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| |
Collapse
|
14
|
Lu SY, Xu QC, Fang DL, Shi YH, Zhu YQ, Liu ZD, Ma MJ, Ye JY, Yin XY. Turning to immunosuppressive tumors: Deciphering the immunosenescence-related microenvironment and prognostic characteristics in pancreatic cancer, in which GLUT1 contributes to gemcitabine resistance. Heliyon 2024; 10:e36684. [PMID: 39263146 PMCID: PMC11388732 DOI: 10.1016/j.heliyon.2024.e36684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024] Open
Abstract
Increasing evidence indicates that the remodeling of immune microenvironment heterogeneity influences pancreatic cancer development, as well as sensitivity to chemotherapy and immunotherapy. However, a gap remains in the exploration of the immunosenescence microenvironment in pancreatic cancer. In this study, we identified two immunosenescence-associated isoforms (IMSP1 and IMSP2), with consequential differences in prognosis and immune cell infiltration. We constructed the MLIRS score, a hazard score system with robust prognostic performance (area under the curve, AUC = 0.91), based on multiple machine learning algorithms (101 cross-validation methods). Patients in the high MLIRS score group had worse prognosis (P < 0.0001) and lower abundance of immune cell infiltration. Conversely, the low MLIRS score group showed better sensitivity to chemotherapy and immunotherapy. Additionally, our MLIRS system outperformed 68 other published signatures. We identified the immunosenescence microenvironmental windsock GLUT1 with certain co-expression properties with immunosenescence markers. We further demonstrated its positive modulation ability of proliferation, migration, and gemcitabine resistance in pancreatic cancer cells. To conclude, our study focused on training of composite machine learning algorithms in multiple datasets to develop a robust machine learning modeling system based on immunosenescence and to identify an immunosenescence-related microenvironment windsock, providing direction and guidance for clinical prediction and application.
Collapse
Affiliation(s)
- Si-Yuan Lu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Qiong-Cong Xu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - De-Liang Fang
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yin-Hao Shi
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ying-Qin Zhu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhi-De Liu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ming-Jian Ma
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Jing-Yuan Ye
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiao Yu Yin
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
15
|
Akmeşe ÖF. Data privacy-aware machine learning approach in pancreatic cancer diagnosis. BMC Med Inform Decis Mak 2024; 24:248. [PMID: 39237927 PMCID: PMC11375871 DOI: 10.1186/s12911-024-02657-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
PROBLEM Pancreatic ductal adenocarcinoma (PDAC) is considered a highly lethal cancer due to its advanced stage diagnosis. The five-year survival rate after diagnosis is less than 10%. However, if diagnosed early, the five-year survival rate can reach up to 70%. Early diagnosis of PDAC can aid treatment and improve survival rates by taking necessary precautions. The challenge is to develop a reliable, data privacy-aware machine learning approach that can accurately diagnose pancreatic cancer with biomarkers. AIM The study aims to diagnose a patient's pancreatic cancer while ensuring the confidentiality of patient records. In addition, the study aims to guide researchers and clinicians in developing innovative methods for diagnosing pancreatic cancer. METHODS Machine learning, a branch of artificial intelligence, can identify patterns by analyzing large datasets. The study pre-processed a dataset containing urine biomarkers with operations such as filling in missing values, cleaning outliers, and feature selection. The data was encrypted using the Fernet encryption algorithm to ensure confidentiality. Ten separate machine learning models were applied to predict individuals with PDAC. Performance metrics such as F1 score, recall, precision, and accuracy were used in the modeling process. RESULTS Among the 590 clinical records analyzed, 199 (33.7%) belonged to patients with pancreatic cancer, 208 (35.3%) to patients with non-cancerous pancreatic disorders (such as benign hepatobiliary disease), and 183 (31%) to healthy individuals. The LGBM algorithm showed the highest efficiency by achieving an accuracy of 98.8%. The accuracy of the other algorithms ranged from 98 to 86%. In order to understand which features are more critical and which data the model is based on, the analysis found that the features "plasma_CA19_9", REG1A, TFF1, and LYVE1 have high importance levels. The LIME analysis also analyzed which features of the model are important in the decision-making process. CONCLUSIONS This research outlines a data privacy-aware machine learning tool for predicting PDAC. The results show that a promising approach can be presented for clinical application. Future research should expand the dataset and focus on validation by applying it to various populations.
Collapse
Affiliation(s)
- Ömer Faruk Akmeşe
- Department of Computer Engineering, Hitit University Çorum, Çorum, 19030, Türkiye.
| |
Collapse
|
16
|
Boekestijn B, Feshtali S, Vasen H, van Leerdam ME, Bonsing BA, Mieog JSD, Wasser MN. Screening for pancreatic cancer in high-risk individuals using MRI: optimization of scan techniques to detect small lesions. Fam Cancer 2024; 23:295-308. [PMID: 38733421 PMCID: PMC11254973 DOI: 10.1007/s10689-024-00394-z] [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: 11/19/2023] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Pancreatic cancer has a dismal prognosis in the general population. However, early detection and treatment of disease in high-risk individuals can improve survival, as patients with localized disease and especially patients with lesions smaller than 10 mm show greatly improved 5-year survival rates. To achieve early detection through MRI surveillance programs, optimization of imaging is required. Advances in MRI technologies in both hardware and software over the years have enabled reliable detection of pancreatic cancer at a small size and early stage. Standardization of dedicated imaging protocols for the pancreas are still lacking. In this review we discuss state of the art scan techniques, sequences, reduction of artifacts and imaging strategies that enable early detection of lesions. Furthermore, we present the imaging features of small pancreatic cancers from a large cohort of high-risk individuals. Refinement of MRI techniques, increased scan quality and the use of artificial intelligence may further improve early detection and the prognosis of pancreatic cancer in a screening setting.
Collapse
Affiliation(s)
- Bas Boekestijn
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Shirin Feshtali
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans Vasen
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin N Wasser
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
Acharjee A, Okyere D, Nath D, Nagar S, Gkoutos GV. Network dynamics and therapeutic aspects of mRNA and protein markers with the recurrence sites of pancreatic cancer. Heliyon 2024; 10:e31437. [PMID: 38803850 PMCID: PMC11128524 DOI: 10.1016/j.heliyon.2024.e31437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease that typically manifests late patient presentation and poor outcomes. Furthermore, PDAC recurrence is a common challenge. Distinct patterns of PDAC recurrence have been associated with differential activation of immune pathway-related genes and specific inflammatory responses in their tumour microenvironment. However, the molecular associations between and within cellular components that underpin PDAC recurrence require further development, especially from a multi-omics integration perspective. In this study, we identified stable molecular associations across multiple PDAC recurrences and utilised integrative analytics to identify stable and novel associations via simultaneous feature selection. Spatial transcriptome and proteome datasets were used to perform univariate analysis, Spearman partial correlation analysis, and univariate analyses by Machine Learning methods, including regularised canonical correlation analysis and sparse partial least squares. Furthermore, networks were constructed for reported and new stable associations. Our findings revealed gene and protein associations across multiple PDAC recurrence groups, which can provide a better understanding of the multi-layer disease mechanisms that contribute to PDAC recurrence. These findings may help to provide novel association targets for clinical studies for constructing precision medicine and personalised surveillance tools for patients with PDAC recurrence.
Collapse
Affiliation(s)
- Animesh Acharjee
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Health Data Research UK (HDR UK), Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, B15 2TT, United Kingdom
- Centre for Health Data Research, University of Birmingham, B15 2TT, United Kingdom
| | - Daniella Okyere
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Dipanwita Nath
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Shruti Nagar
- Eureka Tutorials, Muzaffarnagar, U.P., 251201, India
| | - Georgios V. Gkoutos
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Health Data Research UK (HDR UK), Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, B15 2TT, United Kingdom
- Centre for Health Data Research, University of Birmingham, B15 2TT, United Kingdom
| |
Collapse
|
21
|
Ren S, Qian LC, Cao YY, Daniels MJ, Song LN, Tian Y, Wang ZQ. Computed tomography-based radiomics diagnostic approach for differential diagnosis between early- and late-stage pancreatic ductal adenocarcinoma. World J Gastrointest Oncol 2024; 16:1256-1267. [PMID: 38660647 PMCID: PMC11037050 DOI: 10.4251/wjgo.v16.i4.1256] [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: 11/05/2023] [Revised: 12/27/2023] [Accepted: 02/01/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND One of the primary reasons for the dismal survival rates in pancreatic ductal adenocarcinoma (PDAC) is that most patients are usually diagnosed at late stages. There is an urgent unmet clinical need to identify and develop diagnostic methods that could precisely detect PDAC at its earliest stages. AIM To evaluate the potential value of radiomics analysis in the differentiation of early-stage PDAC from late-stage PDAC. METHODS A total of 71 patients with pathologically proved PDAC based on surgical resection who underwent contrast-enhanced computed tomography (CT) within 30 d prior to surgery were included in the study. Tumor staging was performed in accordance with the 8th edition of the American Joint Committee on Cancer staging system. Radiomics features were extracted from the region of interest (ROI) for each patient using Analysis Kit software. The most important and predictive radiomics features were selected using Mann-Whitney U test, univariate logistic regression analysis, and minimum redundancy maximum relevance (MRMR) method. Random forest (RF) method was used to construct the radiomics model, and 10-times leave group out cross-validation (LGOCV) method was used to validate the robustness and reproducibility of the model. RESULTS A total of 792 radiomics features (396 from late arterial phase and 396 from portal venous phase) were extracted from the ROI for each patient using Analysis Kit software. Nine most important and predictive features were selected using Mann-Whitney U test, univariate logistic regression analysis, and MRMR method. RF method was used to construct the radiomics model with the nine most predictive radiomics features, which showed a high discriminative ability with 97.7% accuracy, 97.6% sensitivity, 97.8% specificity, 98.4% positive predictive value, and 96.8% negative predictive value. The radiomics model was proved to be robust and reproducible using 10-times LGOCV method with an average area under the curve of 0.75 by the average performance of the 10 newly built models. CONCLUSION The radiomics model based on CT could serve as a promising non-invasive method in differential diagnosis between early and late stage PDAC.
Collapse
Affiliation(s)
- Shuai Ren
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Li-Chao Qian
- Department of Geratology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu Province, China
| | - Ying-Ying Cao
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Marcus J Daniels
- Department of Radiology, NYU Langone Health, New York, NY 10016, United States
| | - Li-Na Song
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Ying Tian
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Zhong-Qiu Wang
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
22
|
Campion JR, O'Connor DB, Lahiff C. Human-artificial intelligence interaction in gastrointestinal endoscopy. World J Gastrointest Endosc 2024; 16:126-135. [PMID: 38577646 PMCID: PMC10989254 DOI: 10.4253/wjge.v16.i3.126] [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: 12/31/2023] [Revised: 01/18/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
The number and variety of applications of artificial intelligence (AI) in gastrointestinal (GI) endoscopy is growing rapidly. New technologies based on machine learning (ML) and convolutional neural networks (CNNs) are at various stages of development and deployment to assist patients and endoscopists in preparing for endoscopic procedures, in detection, diagnosis and classification of pathology during endoscopy and in confirmation of key performance indicators. Platforms based on ML and CNNs require regulatory approval as medical devices. Interactions between humans and the technologies we use are complex and are influenced by design, behavioural and psychological elements. Due to the substantial differences between AI and prior technologies, important differences may be expected in how we interact with advice from AI technologies. Human–AI interaction (HAII) may be optimised by developing AI algorithms to minimise false positives and designing platform interfaces to maximise usability. Human factors influencing HAII may include automation bias, alarm fatigue, algorithm aversion, learning effect and deskilling. Each of these areas merits further study in the specific setting of AI applications in GI endoscopy and professional societies should engage to ensure that sufficient emphasis is placed on human-centred design in development of new AI technologies.
Collapse
Affiliation(s)
- John R Campion
- Department of Gastroenterology, Mater Misericordiae University Hospital, Dublin D07 AX57, Ireland
- School of Medicine, University College Dublin, Dublin D04 C7X2, Ireland
| | - Donal B O'Connor
- Department of Surgery, Trinity College Dublin, Dublin D02 R590, Ireland
| | - Conor Lahiff
- Department of Gastroenterology, Mater Misericordiae University Hospital, Dublin D07 AX57, Ireland
- School of Medicine, University College Dublin, Dublin D04 C7X2, Ireland
| |
Collapse
|
23
|
Ahmed HS. BEYOND TRADITIONAL TOOLS: EXPLORING CONVOLUTIONAL NEURAL NETWORKS AS INNOVATIVE PROGNOSTIC MODELS IN PANCREATIC DUCTAL ADENOCARCINOMA. ARQUIVOS DE GASTROENTEROLOGIA 2024; 61:e23107. [PMID: 38511794 DOI: 10.1590/s0004-2803.24612023-117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 02/07/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive and lethal form of cancer with limited prognostic accuracy using traditional factors. This has led to the exploration of innovative prognostic models, including convolutional neural networks (CNNs), in PDAC. CNNs, a type of artificial intelligence algorithm, have shown promise in various medical applications, including image analysis and pattern recognition. Their ability to extract complex features from medical images makes them suitable for improving prognostication in PDAC. However, implementing CNNs in clinical practice poses challenges, such as data availability and interpretability. Future research should focus on multi-center studies, integrating multiple data modalities, and combining CNN outputs with biomarker panels. Collaborative efforts and patient autonomy should be considered to ensure the ethical implementation of CNN-based prognostic models. Further validation and optimisation of CNN-based models are necessary to enhance their reliability and clinical utility in PDAC prognostication. BACKGROUND •Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with limited prognostic accuracy through traditional methods. BACKGROUND •Convolutional neural networks (CNNs) are being explored for prognostic models in PDAC. BACKGROUND •They can extract complex features from images, aiding PDAC prognostication. BACKGROUND •Further validation and optimization of CNN-based models are needed for better reliability and clinical utility in PDAC.
Collapse
|
24
|
Wen YR, Lin XW, Zhou YW, Xu L, Zhang JL, Chen CY, He J. N-glycan biosignatures as a potential diagnostic biomarker for early-stage pancreatic cancer. World J Gastrointest Oncol 2024; 16:659-669. [PMID: 38577461 PMCID: PMC10989390 DOI: 10.4251/wjgo.v16.i3.659] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, with a 5-year survival rate of less than 10%, owing to its late-stage diagnosis. Early detection of pancreatic cancer (PC) can significantly increase survival rates. AIM To identify the serum biomarker signatures associated with early-stage PDAC by serum N-glycan analysis. METHODS An extensive patient cohort was used to determine a biomarker signature, including patients with PDAC that was well-defined at an early stage (stages I and II). The biomarker signature was derived from a case-control study using a case-cohort design consisting of 29 patients with stage I, 22 with stage II, 4 with stage III, 16 with stage IV PDAC, and 88 controls. We used multiparametric analysis to identify early-stage PDAC N-glycan signatures and developed an N-glycan signature-based diagnosis model called the "Glyco-model". RESULTS The biomarker signature was created to discriminate samples derived from patients with PC from those of controls, with a receiver operating characteristic area under the curve of 0.86. In addition, the biomarker signature combined with cancer antigen 19-9 could discriminate patients with PDAC from controls, with a receiver operating characteristic area under the curve of 0.919. Glyco-model demonstrated favorable diagnostic performance in all stages of PC. The diagnostic sensitivity for stage I PDAC was 89.66%. CONCLUSION In a prospective validation study, this serum biomarker signature may offer a viable method for detecting early-stage PDAC.
Collapse
Affiliation(s)
- Yan-Rong Wen
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Xia-Wen Lin
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Yu-Wen Zhou
- Department of Research and Development, Sysdiagno (Nanjing) Biotech Co., Ltd, Nanjing 210008, Jiangsu Province, China
| | - Lei Xu
- Department of Research and Development, Sysdiagno (Nanjing) Biotech Co., Ltd, Nanjing 210008, Jiangsu Province, China
| | - Jun-Li Zhang
- Department of Research and Development, Sysdiagno (Nanjing) Biotech Co., Ltd, Nanjing 210008, Jiangsu Province, China
| | - Cui-Ying Chen
- Department of Research and Development, Sysdiagno (Nanjing) Biotech Co., Ltd, Nanjing 210008, Jiangsu Province, China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
| |
Collapse
|
25
|
Huma C, Hawon L, Sarisha J, Erdal T, Kevin C, Valentina KA. Advances in the field of developing biomarkers for re-irradiation: a how-to guide to small, powerful data sets and artificial intelligence. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2024; 9:3-16. [PMID: 38550554 PMCID: PMC10972602 DOI: 10.1080/23808993.2024.2325936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/28/2024] [Indexed: 04/01/2024]
Abstract
Introduction Patient selection remains challenging as the clinical use of re-irradiation (re-RT) increases. Re-RT data is limited to retrospective studies and small prospective single-institution reports, resulting in small, heterogenous data sets. Validated prognostic and predictive biomarkers are derived from large-volume studies with long-term follow-up. This review aims to examine existing re-RT publications and available data sets and discuss strategies using artificial intelligence (AI) to approach small data sets to optimize the use of re-RT data. Methods Re-RT publications were identified where associated public data was present. The existing literature on small data sets to identify biomarkers was also explored. Results Publications with associated public data were identified, with glioma and nasopharyngeal cancers emerging as the most common tumor sites where the use of re-RT was the primary management approach. Existing and emerging AI strategies have been used to approach small data sets including data generation, augmentation, discovery, and transfer learning. Conclusions Further data is needed to generate adaptive frameworks, improve the collection of specimens for molecular analysis, and improve the interpretability of results in re-RT data.
Collapse
Affiliation(s)
- Chaudhry Huma
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| | - Lee Hawon
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| | - Jagasia Sarisha
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| | - Tasci Erdal
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| | - Camphausen Kevin
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| | - Krauze Andra Valentina
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Bethesda, MD, 20892, United States
| |
Collapse
|
26
|
Daher H, Punchayil SA, Ismail AAE, Fernandes RR, Jacob J, Algazzar MH, Mansour M. Advancements in Pancreatic Cancer Detection: Integrating Biomarkers, Imaging Technologies, and Machine Learning for Early Diagnosis. Cureus 2024; 16:e56583. [PMID: 38646386 PMCID: PMC11031195 DOI: 10.7759/cureus.56583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 04/23/2024] Open
Abstract
Artificial intelligence (AI) has come to play a pivotal role in revolutionizing medical practices, particularly in the field of pancreatic cancer detection and management. As a leading cause of cancer-related deaths, pancreatic cancer warrants innovative approaches due to its typically advanced stage at diagnosis and dismal survival rates. Present detection methods, constrained by limitations in accuracy and efficiency, underscore the necessity for novel solutions. AI-driven methodologies present promising avenues for enhancing early detection and prognosis forecasting. Through the analysis of imaging data, biomarker profiles, and clinical information, AI algorithms excel in discerning subtle abnormalities indicative of pancreatic cancer with remarkable precision. Moreover, machine learning (ML) algorithms facilitate the amalgamation of diverse data sources to optimize patient care. However, despite its huge potential, the implementation of AI in pancreatic cancer detection faces various challenges. Issues such as the scarcity of comprehensive datasets, biases in algorithm development, and concerns regarding data privacy and security necessitate thorough scrutiny. While AI offers immense promise in transforming pancreatic cancer detection and management, ongoing research and collaborative efforts are indispensable in overcoming technical hurdles and ethical dilemmas. This review delves into the evolution of AI, its application in pancreatic cancer detection, and the challenges and ethical considerations inherent in its integration.
Collapse
Affiliation(s)
- Hisham Daher
- Internal Medicine, University of Debrecen, Debrecen, HUN
| | - Sneha A Punchayil
- Internal Medicine, University Hospital of North Tees, Stockton-on-Tees, GBR
| | | | | | - Joel Jacob
- General Medicine, Diana Princess of Wales Hospital, Grimsby, GBR
| | | | - Mohammad Mansour
- General Medicine, University of Debrecen, Debrecen, HUN
- General Medicine, Jordan University Hospital, Amman, JOR
| |
Collapse
|
27
|
Marchese U, Rebours V, Sauvanet A, Caron O, Ali EA, Perkins G, Malka D, Dohan A, Thibault LM, Perrod G, Buecher B. [Hereditary and familial forms of pancreatic adenocarcinoma: Genetic determinism, patients eligible for systematic screening, screening methods and results]. Bull Cancer 2024; 111:199-212. [PMID: 38123413 DOI: 10.1016/j.bulcan.2023.11.002] [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: 08/31/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023]
Abstract
Systematic screening for pancreatic cancer in high risk individuals is justified by the poor prognosis of the majority of cases diagnosed at a symptomatic stage that are mostly advanced and unresectable Individual risk assessment is based on both genetic data and family history. The screening of a panel of susceptibiility genes should be offered to any affected individual when a genetic predisposition is suspected. An international consortium has proposed a definition of the at risk population, candidate for screening, and there is a consensus on the target lesions of this screening: early adenocarcinoma and benign lesions with a high potential for malignant transformation: Intraductal Papillary Mucinous Neopasm (IPMN) and Pancreatic Intraepithelial Neoplasia (PanIN) with high-grade dysplasia. Its modalities currently consist of an annual pancreatic MRI and/or endoscopic ultrasound (EUS), associated with screening for diabetes mellitus. The main limitation of screening, the effectiveness of which has not yet been demonstrated, is its lack of sensitivity, which results in a non-negligible rate of interval cancers and sometimes advanced diagnoses. Insufficient specificity is also imperfect, in particular with regard to benign lesions with a low potential for degeneration, and can lead to the proposal of unjustified surgeries. This situation makes the future integration of new imaging techniques and promising new biological approaches that are being explored highly desirable.
Collapse
Affiliation(s)
- Ugo Marchese
- AP-HP, hôpital Cochin, université de Paris, centre, service de chirurgie digestive, hépatobiliaire et endocrinienne, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Vinciane Rebours
- AP-HP, hôpital Beaujon - Clichy, université Paris-Cité, service de pancréatologie et oncologie digestive, 100, boulevard du Général Leclerc, 92110 Clichy, France
| | - Alain Sauvanet
- AP-HP, hôpital Beaujon - Clichy, université Paris-Cité, département chirurgie hépato-biliaire et pancréatique, 100, boulevard du Général Leclerc, 92110 Clichy, France
| | - Olivier Caron
- Gustave-Roussy, département de médecine oncologique, 94805 Villejuif, France
| | - Einas Abou Ali
- AP-HP, hôpital Cochin, université de Paris, centre, service de gastro-entérologie et oncologie digestive, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Géraldine Perkins
- AP-HP, hôpital européen Georges-Pompidou, Centre, université Paris-Cité, unité d'oncogénétique, 20, rue Leblanc, 75015 Paris, France
| | - David Malka
- Institut mutualiste Montsouris, département d'oncologie médicale, 42, boulevard Jourdan, 75014 Paris, France
| | - Anthony Dohan
- AP-HP, hôpital Cochin, université de Paris, centre, service de radiologie, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Louise May Thibault
- Centre François-Baclesse, service d'oncogénétique, unité de biopathologie, 3, avenue de Général Harris, 14000 Caen, France
| | - Guillaume Perrod
- AP-HP, hôpital européen Georges-Pompidou, université Paris-Cité, centre, département d'hépato-gastroentérologie et endoscopies digestives, 20, rue Leblanc, 75015 Paris, France
| | - Bruno Buecher
- PSL Research University, institut Curie, service de génétique, pôle médecine diagnostique & théranostique, 26, rue d'Ulm, 75248 Paris cédex 05, France.
| |
Collapse
|
28
|
Yimamu A, Li J, Zhang H, Liang L, Feng L, Wang Y, Zhou C, Li S, Gao Y. Computed tomography and guidelines-based human-machine fusion model for predicting resectability of the pancreatic cancer. J Gastroenterol Hepatol 2024; 39:399-409. [PMID: 37957952 DOI: 10.1111/jgh.16401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/04/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND AND AIM The study aims to develop a hybrid machine learning model for predicting resectability of the pancreatic cancer, which is based on computed tomography (CT) and National Comprehensive Cancer Network (NCCN) guidelines. METHOD We retrospectively studied 349 patients. One hundred seventy-one cases from Center 1 and 92 cases from Center 2 were used as the primary training cohort, and 66 cases from Center 3 and 20 cases from Center 4 were used as the independent test dataset. Semi-automatic module of ITK-SNAP software was used to assist CT image segmentation to obtain three-dimensional (3D) imaging region of interest (ROI). There were 788 handcrafted features extracted for 3D ROI using PyRadiomics. The optimal feature subset consists of three features screened by three feature selection methods as the input of the SVM to construct the conventional radiomics-based predictive model (cRad). 3D ROI was used to unify the resolution by 3D spline interpolation method for constructing the 3D tumor imaging tensor. Using 3D tumor image tensor as input, 3D kernelled support tensor machine-based predictive model (KSTM), and 3D ResNet-based deep learning predictive model (ResNet) were constructed. Multi-classifier fusion ML model is constructed by fusing cRad, KSTM, and ResNet using multi-classifier fusion strategy. Two experts with more than 10 years of clinical experience were invited to reevaluate each patient based on their CECT following the NCCN guidelines to obtain resectable, unresectable, and borderline resectable diagnoses. The three results were converted into probability values of 0.25, 0.75, and 0.50, respectively, according to the traditional empirical method. Then it is used as an independent classifier and integrated with multi-classifier fusion machine learning (ML) model to obtain the human-machine fusion ML model (HMfML). RESULTS Multi-classifier fusion ML model's area under receiver operating characteristic curve (AUC; 0.8610), predictive accuracy (ACC: 80.23%), sensitivity (SEN: 78.95%), and specificity (SPE: 80.60%) is better than cRad, KSTM, and ResNet-based single-classifier models and their two-classifier fusion models. This means that three different models have mined complementary CECT feature expression from different perspectives and can be integrated through CFS-ER, so that the fusion model has better performance. HMfML's AUC (0.8845), ACC (82.56%), SEN (84.21%), SPE (82.09%). This means that ML models might learn extra information from CECT that experts cannot distinguish, thus complementing expert experience and improving the performance of hybrid ML models. CONCLUSION HMfML can predict PC resectability with high accuracy.
Collapse
Affiliation(s)
- Adilijiang Yimamu
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Li
- School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medical Image Processing, Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
| | - Haojie Zhang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lidu Liang
- School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medical Image Processing, Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
| | - Lei Feng
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Wang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chenjie Zhou
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shulong Li
- School of Biomedical Engineering, Guangdong Provincial Key Laboratory of Medical Image Processing, Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
| | - Yi Gao
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
29
|
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
|
30
|
Villani A, Fontana A, Panebianco C, Ferro C, Copetti M, Pavlovic R, Drago D, Fiorentini C, Terracciano F, Bazzocchi F, Canistro G, Pisati F, Maiello E, Latiano TP, Perri F, Pazienza V. A powerful machine learning approach to identify interactions of differentially abundant gut microbial subsets in patients with metastatic and non-metastatic pancreatic cancer. Gut Microbes 2024; 16:2375483. [PMID: 38972056 PMCID: PMC11229760 DOI: 10.1080/19490976.2024.2375483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 06/28/2024] [Indexed: 07/09/2024] Open
Abstract
Pancreatic cancer has a dismal prognosis, as it is often diagnosed at stage IV of the disease and is characterized by metastatic spread. Gut microbiota and its metabolites have been suggested to influence the metastatic spread by modulating the host immune system or by promoting angiogenesis. To date, the gut microbial profiles of metastatic and non-metastatic patients need to be explored. Taking advantage of the 16S metagenomic sequencing and the PEnalized LOgistic Regression Analysis (PELORA) we identified clusters of bacteria with differential abundances between metastatic and non-metastatic patients. An overall increase in Gram-negative bacteria in metastatic patients compared to non-metastatic ones was identified using this method. Furthermore, to gain more insight into how gut microbes can predict metastases, a machine learning approach (iterative Random Forest) was performed. Iterative Random Forest analysis revealed which microorganisms were characterized by a different level of relative abundance between metastatic and non-metastatic patients and established a functional relationship between the relative abundance and the probability of having metastases. At the species level, the following bacteria were found to have the highest discriminatory power: Anaerostipes hadrus, Coprobacter secundus, Clostridium sp. 619, Roseburia inulinivorans, Porphyromonas and Odoribacter at the genus level, and Rhodospirillaceae, Clostridiaceae and Peptococcaceae at the family level. Finally, these data were intertwined with those from a metabolomics analysis on fecal samples of patients with or without metastasis to better understand the role of gut microbiota in the metastatic process. Artificial intelligence has been applied in different areas of the medical field. Translating its application in the field of gut microbiota analysis may help fully exploit the potential information contained in such a large amount of data aiming to open up new supportive areas of intervention in the management of cancer.
Collapse
Affiliation(s)
- Annacandida Villani
- Division of Gastroenterology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Andrea Fontana
- Biostatistic Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Concetta Panebianco
- Division of Gastroenterology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Carmelapia Ferro
- Division of Gastroenterology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Massimiliano Copetti
- Biostatistic Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Radmila Pavlovic
- Proteomics and Metabolomics Facility (ProMeFa), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Denise Drago
- Proteomics and Metabolomics Facility (ProMeFa), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carla Fiorentini
- Scientific Direction, Association for Research on Integrative Oncological Therapies (ARTOI), Roma, Italy
| | - Fulvia Terracciano
- Division of Gastroenterology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Francesca Bazzocchi
- Abdominal Surgery Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Giuseppe Canistro
- Abdominal Surgery Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | | | - Evaristo Maiello
- Oncology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Tiziana Pia Latiano
- Oncology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Francesco Perri
- Division of Gastroenterology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Valerio Pazienza
- Division of Gastroenterology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
31
|
Flammia F, Fusco R, Triggiani S, Pellegrino G, Reginelli A, Simonetti I, Trovato P, Setola SV, Petralia G, Petrillo A, Izzo F, Granata V. Risk Assessment and Radiomics Analysis in Magnetic Resonance Imaging of Pancreatic Intraductal Papillary Mucinous Neoplasms (IPMN). Cancer Control 2024; 31:10732748241263644. [PMID: 39293798 PMCID: PMC11412216 DOI: 10.1177/10732748241263644] [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: 09/20/2024] Open
Abstract
Intraductal papillary mucinous neoplasms (IPMNs) are a very common incidental finding during patient radiological assessment. These lesions may progress from low-grade dysplasia (LGD) to high-grade dysplasia (HGD) and even pancreatic cancer. The IPMN progression risk grows with time, so discontinuation of surveillance is not recommended. It is very important to identify imaging features that suggest LGD of IPMNs, and thus, distinguish lesions that only require careful surveillance from those that need surgical resection. It is important to know the management guidelines and especially the indications for surgery, to be able to point out in the report the findings that suggest malignant degeneration. The imaging tools employed for diagnosis and risk assessment are Computed Tomography (CT) and Magnetic Resonance Imaging (MRI) with contrast medium. According to the latest European guidelines, MRI is the method of choice for the diagnosis and follow-up of patients with IPMN since this tool has a highest sensitivity in detecting mural nodules and intra-cystic septa. It plays a key role in the diagnosis of worrisome features and high-risk stigmata, which are associated with IPMNs malignant degeneration. Nowadays, the main limit of diagnostic tools is the ability to identify the precursor of pancreatic cancer. In this context, increasing attention is being given to artificial intelligence (AI) and radiomics analysis. However, these tools remain in an exploratory phase, considering the limitations of currently published studies. Key limits include noncompliance with AI best practices, radiomics workflow standardization, and clear reporting of study methodology, including segmentation and data balancing. In the radiological report it is useful to note the type of IPMN so as the morphological features, size, rate growth, wall, septa and mural nodules, on which the indications for surveillance and surgery are based. These features should be reported so as the surveillance time should be suggested according to guidelines.
Collapse
Affiliation(s)
- Federica Flammia
- SIRM Foundation, Italian Society of Medical and Interventional Radiology (SIRM), Milan, Italy
| | | | - Sonia Triggiani
- Postgraduate School of Radiodiagnostics, University of Milan, Milan, Italy
| | | | - Alfonso Reginelli
- Division of Radiology, "Università Degli Studi Della Campania Luigi Vanvitelli", Naples, Italy
| | - Igino Simonetti
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Piero Trovato
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Sergio Venanzio Setola
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Giuseppe Petralia
- Radiology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
- Departement of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Antonella Petrillo
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Francesco Izzo
- Divisions of Hepatobiliary Surgery, "Istituto Nazionale dei Tumori IRCCS Fondazione G. Pascale", Naples, Italy
| | - Vincenza Granata
- Radiology Division, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
32
|
Panagiotidis E, Papachristou K, Makridou A, Zoglopitou LA, Paschali A, Kalathas T, Chatzimarkou M, Chatzipavlidou V. Review of artificial intelligence clinical applications in Nuclear Medicine. Nucl Med Commun 2024; 45:24-34. [PMID: 37901920 DOI: 10.1097/mnm.0000000000001786] [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: 10/31/2023]
Abstract
This paper provides an in-depth analysis of the clinical applications of artificial intelligence (AI) in Nuclear Medicine, focusing on three key areas: neurology, cardiology, and oncology. Beginning with neurology, specifically Alzheimer's disease and Parkinson's disease, the paper examines reviews on diagnosis and treatment planning. The same pattern is followed in cardiology studies. In the final section on oncology, the paper explores the various AI applications in multiple cancer types, including lung, head and neck, lymphoma, and pancreatic cancer.
Collapse
Affiliation(s)
| | | | - Anna Makridou
- Medical Physics Department, Cancer Hospital of Thessaloniki 'Theagenio', Thessaloniki, Greece
| | | | - Anna Paschali
- Nuclear Medicine Department, Cancer Hospital of Thessaloniki 'Theagenio' and
| | - Theodoros Kalathas
- Nuclear Medicine Department, Cancer Hospital of Thessaloniki 'Theagenio' and
| | - Michael Chatzimarkou
- Medical Physics Department, Cancer Hospital of Thessaloniki 'Theagenio', Thessaloniki, Greece
| | | |
Collapse
|
33
|
Berbís MÁ, Godino FP, Rodríguez-Comas J, Nava E, García-Figueiras R, Baleato-González S, Luna A. Radiomics in CT and MR imaging of the liver and pancreas: tools with potential for clinical application. Abdom Radiol (NY) 2024; 49:322-340. [PMID: 37889265 DOI: 10.1007/s00261-023-04071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023]
Abstract
Radiomics allows the extraction of quantitative imaging features from clinical magnetic resonance imaging (MRI) and computerized tomography (CT) studies. The advantages of radiomics have primarily been exploited in oncological applications, including better characterization and staging of oncological lesions and prediction of patient outcomes and treatment response. The potential introduction of radiomics in the clinical setting requires the establishment of a standardized radiomics pipeline and a quality assurance program. Radiomics and texture analysis of the liver have improved the differentiation of hypervascular lesions such as adenomas, focal nodular hyperplasia, and hepatocellular carcinoma (HCC) during the arterial phase, and in the pretreatment determination of HCC prognostic factors (e.g., tumor grade, microvascular invasion, Ki-67 proliferation index). Radiomics of pancreatic CT and MR images has enhanced pancreatic ductal adenocarcinoma detection and its differentiation from pancreatic neuroendocrine tumors, mass-forming chronic pancreatitis, or autoimmune pancreatitis. Radiomics can further help to better characterize incidental pancreatic cystic lesions, accurately discriminating benign from malignant intrapancreatic mucinous neoplasms. Nonetheless, despite their encouraging results and exciting potential, these tools have yet to be implemented in the clinical setting. This non-systematic review will describe the essential steps in the implementation of the radiomics and feature extraction workflow from liver and pancreas CT and MRI studies for their potential clinical application. A succinct overview of reported radiomics applications in the liver and pancreas and the challenges and limitations of their implementation in the clinical setting is also discussed, concluding with a brief exploration of the future perspectives of radiomics in the gastroenterology field.
Collapse
Affiliation(s)
- M Álvaro Berbís
- Department of Radiology, HT Médica, San Juan de Dios Hospital, 14960, Córdoba, Spain.
- Department of Radiology, HT Médica, San Juan de Dios Hospital, Av. del Brillante, 106, 14012, Córdoba, Spain.
| | | | | | - Enrique Nava
- Department of Communications Engineering, University of Málaga, 29016, Málaga, Spain
| | - Roberto García-Figueiras
- Abdominal Imaging Section, University Clinical Hospital of Santiago, 15706, Santiago de Compostela, A Coruña, Spain
| | - Sandra Baleato-González
- Abdominal Imaging Section, University Clinical Hospital of Santiago, 15706, Santiago de Compostela, A Coruña, Spain
| | - Antonio Luna
- Department of Radiology, HT Médica, Clínica las Nieves, 23007, Jaén, Spain
| |
Collapse
|
34
|
Quagliarini E, Caputo D, Cammarata R, Caracciolo G, Pozzi D. Coupling magnetic levitation of graphene oxide–protein complexes with blood levels of glucose for early detection of pancreatic adenocarcinoma. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00170-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Abstract
Introduction
Pancreatic adenocarcinoma (PDAC) has a poor prognosis since often diagnosed too late. Dyslipidemia and hyperglycemia are considered risk factors, but the presence of the tumor itself can determine the onset of these disorders. Therefore, it is not easy to predict which subjects with diabetes or dyslipidemia will develop or have already developed PDAC. Over the past decade, tests based on the use of nanotechnology, alone or coupled with common laboratory tests (e.g., hemoglobin levels), have proven useful in aiding the diagnosis of PDAC. Tests based on magnetic levitation (MagLev) have demonstrated high diagnostic accuracy in compliance with the REASSURED criteria. Here, we aimed to assess the ability of the MagLev test in detecting PDAC when coupled with the blood levels of glycemia, cholesterol, and triglycerides.
Methods
Blood samples from 24 PDAC patients and 22 healthy controls were collected. Human plasma was let to interact with graphene oxide (GO) nanosheets and the emerging coronated systems were put in the MagLev device. Outcomes from Maglev experiments were coupled to glycemia, cholesterol, and triglycerides levels. Linear discriminant analysis (LDA) was carried out to evaluate the classification ability of the test in terms of specificity, sensitivity, and global accuracy. Statistical analysis was performed with Matlab (MathWorks, Natick, MA, USA, Version R2022a) software.
Results
The positions of the levitating bands were measured at the starting point (i.e., as soon as the cuvette containing the sample was subjected to the magnetic field). Significant variations in the starting position of levitating nanosystems in controls and PDACs were detected. The combination of the MagLev outcomes with the blood glycemic levels returned the best value of global accuracy (91%) if compared to the coupling with those of cholesterol and triglycerides (global accuracy of ~ 77% and 84%, respectively).
Conclusion
If confirmed by further studies on larger cohorts, a multiplexed Maglev-based nanotechnology-enabled blood test could be employed as a screening tool for PDAC in populations with hyperglycemia.
Collapse
|
35
|
Wojtara M, Rana E, Rahman T, Khanna P, Singh H. Artificial intelligence in rare disease diagnosis and treatment. Clin Transl Sci 2023; 16:2106-2111. [PMID: 37646577 PMCID: PMC10651639 DOI: 10.1111/cts.13619] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/30/2023] [Accepted: 08/13/2023] [Indexed: 09/01/2023] Open
Abstract
Artificial intelligence (AI) utilization in health care has grown over the past few years. It also has demonstrated potential in improving the efficiency of diagnosis and treatment. Some types of AI, such as machine learning, allow for the efficient analysis of vast datasets, identifying patterns, and generating key insights. Predictions can then be made for medical diagnosis and personalized treatment recommendations. The use of AI can bypass some conventional limitations associated with rare diseases. Namely, it can optimize traditional randomized control trials, and may eventually reduce costs for drug research and development. Recent advancements have enabled researchers to train models based on large datasets and then fine-tune these models on smaller datasets typically associated with rare diseases. In this mini-review, we discuss recent advancements in AI and how AI can be applied to streamline rare disease diagnosis and optimize treatment.
Collapse
Affiliation(s)
- Magda Wojtara
- Department of Human GeneticsUniversity of MichiganAnn ArborMichiganUSA
| | - Emaan Rana
- Department of ScienceUniversity of Western OntarioLondonOntarioCanada
| | - Taibia Rahman
- Department of MedicineDavid Tvildiani Medical UniversityTbilisiGeorgia
| | - Palak Khanna
- Department of MedicineIvane Javakhishvili Tbilisi State UniversityTbilisiGeorgia
| | - Heshwin Singh
- Department of BiologyStony Brook UniversityStony BrookNew YorkUSA
| |
Collapse
|
36
|
Caputo D, Quagliarini E, Coppola A, La Vaccara V, Marmiroli B, Sartori B, Caracciolo G, Pozzi D. Inflammatory biomarkers and nanotechnology: new insights in pancreatic cancer early detection. Int J Surg 2023; 109:2934-2940. [PMID: 37352522 PMCID: PMC10583897 DOI: 10.1097/js9.0000000000000558] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/02/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND Poor prognosis of pancreatic ductal adenocarcinoma (PDAC) is mainly due to the lack of effective early-stage detection strategies. Even though the link between inflammation and PDAC has been demonstrated and inflammatory biomarkers proved their efficacy in predicting several tumours, to date they have a role only in assessing PDAC prognosis. Recently, the studies of interactions between nanosystems and easily collectable biological fluids, alone or coupled with standard laboratory tests, have proven useful in facilitating PDAC diagnosis. Notably, tests based on magnetic levitation (MagLev) of biocoronated nanosystems have demonstrated high diagnostic accuracy in compliance with the criteria stated by WHO. Herein, the author developed a synergistic analysis that combines a user-friendly MagLev-based approach and common inflammatory biomarkers for discriminating PDAC subjects from healthy ones. MATERIALS AND METHODS Plasma samples from 24 PDAC subjects and 22 non-oncological patients have been collected and let to interact with graphene oxide nanosheets.Biomolecular corona formed around graphene oxide nanosheets have been immersed in a Maglev platform to study the levitation profiles.Inflammatory biomarkers such as neutrophil-to-lymphocyte ratio (NLR), derived-NLR (dNLR), and platelet to lymphocyte ratio have been calculated and combined with results obtained by the MagLev platform. RESULTS MagLev profiles resulted significantly different between non-oncological patients and PDAC and allowed to identify a MagLev fingerprint for PDAC. Four inflammatory markers were significantly higher in PDAC subjects: neutrophils ( P =0.04), NLR ( P =4.7 ×10 -6 ), dNLR ( P =2.7 ×10 -5 ), and platelet to lymphocyte ratio ( P =0.002). Lymphocytes were appreciably lower in PDACs ( P =2.6 ×10 -6 ).Combining the MagLev fingerprint with dNLR and NLR returned global discrimination accuracy for PDAC of 95.7% and 91.3%, respectively. CONCLUSIONS The multiplexed approach discriminated PDAC patients from healthy volunteers in up to 95% of cases. If further confirmed in larger-cohort studies, this approach may be used for PDAC detection.
Collapse
Affiliation(s)
- Damiano Caputo
- Research Unit of Generale Surgery, Department of Medicine and Surgery, University Campus Bio-Medico di Roma
- Operative Research Unit of General Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo
| | | | - Alessandro Coppola
- Department of Surgery, Sapienza University of Rome, Viale Regina Elena, Rome, Italy
| | - Vincenzo La Vaccara
- Operative Research Unit of General Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo
| | - Benedetta Marmiroli
- Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse, Graz, Austria
| | - Barbara Sartori
- Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse, Graz, Austria
| | | | | |
Collapse
|
37
|
Borna S, Maniaci MJ, Haider CR, Maita KC, Torres-Guzman RA, Avila FR, Lunde JJ, Coffey JD, Demaerschalk BM, Forte AJ. Artificial Intelligence Models in Health Information Exchange: A Systematic Review of Clinical Implications. Healthcare (Basel) 2023; 11:2584. [PMID: 37761781 PMCID: PMC10531020 DOI: 10.3390/healthcare11182584] [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: 08/07/2023] [Revised: 09/14/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
Electronic health record (EHR) systems collate patient data, and the integration and standardization of documents through Health Information Exchange (HIE) play a pivotal role in refining patient management. Although the clinical implications of AI in EHR systems have been extensively analyzed, its application in HIE as a crucial source of patient data is less explored. Addressing this gap, our systematic review delves into utilizing AI models in HIE, gauging their predictive prowess and potential limitations. Employing databases such as Scopus, CINAHL, Google Scholar, PubMed/Medline, and Web of Science and adhering to the PRISMA guidelines, we unearthed 1021 publications. Of these, 11 were shortlisted for the final analysis. A noticeable preference for machine learning models in prognosticating clinical results, notably in oncology and cardiac failures, was evident. The metrics displayed AUC values ranging between 61% and 99.91%. Sensitivity metrics spanned from 12% to 96.50%, specificity from 76.30% to 98.80%, positive predictive values varied from 83.70% to 94.10%, and negative predictive values between 94.10% and 99.10%. Despite variations in specific metrics, AI models drawing on HIE data unfailingly showcased commendable predictive proficiency in clinical verdicts, emphasizing the transformative potential of melding AI with HIE. However, variations in sensitivity highlight underlying challenges. As healthcare's path becomes more enmeshed with AI, a well-rounded, enlightened approach is pivotal to guarantee the delivery of trustworthy and effective AI-augmented healthcare solutions.
Collapse
Affiliation(s)
- Sahar Borna
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Michael J. Maniaci
- Division of Hospital Internal Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Clifton R. Haider
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA
| | - Karla C. Maita
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | | | - Jordan D. Coffey
- Center for Digital Health, Mayo Clinic, Rochester, MN 55902, USA
| | - Bart M. Demaerschalk
- Center for Digital Health, Mayo Clinic, Rochester, MN 55902, USA
- Department of Neurology, Mayo Clinic College of Medicine and Science, Phoenix, AZ 85054, USA
| | - Antonio J. Forte
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
38
|
Kaczmarzyk JR, Gupta R, Kurc TM, Abousamra S, Saltz JH, Koo PK. ChampKit: A framework for rapid evaluation of deep neural networks for patch-based histopathology classification. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 239:107631. [PMID: 37271050 PMCID: PMC11093625 DOI: 10.1016/j.cmpb.2023.107631] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/23/2023] [Accepted: 05/28/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND AND OBJECTIVE Histopathology is the gold standard for diagnosis of many cancers. Recent advances in computer vision, specifically deep learning, have facilitated the analysis of histopathology images for many tasks, including the detection of immune cells and microsatellite instability. However, it remains difficult to identify optimal models and training configurations for different histopathology classification tasks due to the abundance of available architectures and the lack of systematic evaluations. Our objective in this work is to present a software tool that addresses this need and enables robust, systematic evaluation of neural network models for patch classification in histology in a light-weight, easy-to-use package for both algorithm developers and biomedical researchers. METHODS Here we present ChampKit (Comprehensive Histopathology Assessment of Model Predictions toolKit): an extensible, fully reproducible evaluation toolkit that is a one-stop-shop to train and evaluate deep neural networks for patch classification. ChampKit curates a broad range of public datasets. It enables training and evaluation of models supported by timm directly from the command line, without the need for users to write any code. External models are enabled through a straightforward API and minimal coding. As a result, Champkit facilitates the evaluation of existing and new models and deep learning architectures on pathology datasets, making it more accessible to the broader scientific community. To demonstrate the utility of ChampKit, we establish baseline performance for a subset of possible models that could be employed with ChampKit, focusing on several popular deep learning models, namely ResNet18, ResNet50, and R26-ViT, a hybrid vision transformer. In addition, we compare each model trained either from random weight initialization or with transfer learning from ImageNet pretrained models. For ResNet18, we also consider transfer learning from a self-supervised pretrained model. RESULTS The main result of this paper is the ChampKit software. Using ChampKit, we were able to systemically evaluate multiple neural networks across six datasets. We observed mixed results when evaluating the benefits of pretraining versus random intialization, with no clear benefit except in the low data regime, where transfer learning was found to be beneficial. Surprisingly, we found that transfer learning from self-supervised weights rarely improved performance, which is counter to other areas of computer vision. CONCLUSIONS Choosing the right model for a given digital pathology dataset is nontrivial. ChampKit provides a valuable tool to fill this gap by enabling the evaluation of hundreds of existing (or user-defined) deep learning models across a variety of pathology tasks. Source code and data for the tool are freely accessible at https://github.com/SBU-BMI/champkit.
Collapse
Affiliation(s)
- Jakub R Kaczmarzyk
- Department of Biomedical Informatics, Stony Brook Medicine, 101 Nicolls Rd, Stony Brook, 11794, NY, USA; Simons Center for Quantitative Biology, 1 Bungtown Rd, Cold Spring Harbor, 11724, NY, USA.
| | - Rajarsi Gupta
- Department of Biomedical Informatics, Stony Brook Medicine, 101 Nicolls Rd, Stony Brook, 11794, NY, USA
| | - Tahsin M Kurc
- Department of Biomedical Informatics, Stony Brook Medicine, 101 Nicolls Rd, Stony Brook, 11794, NY, USA
| | - Shahira Abousamra
- Department of Computer Science, Stony Brook University, Stony Brook, NY, USA
| | - Joel H Saltz
- Department of Biomedical Informatics, Stony Brook Medicine, 101 Nicolls Rd, Stony Brook, 11794, NY, USA.
| | - Peter K Koo
- Simons Center for Quantitative Biology, 1 Bungtown Rd, Cold Spring Harbor, 11724, NY, USA.
| |
Collapse
|
39
|
Deng S, Li C, Cao J, Cui Z, Du J, Fu Z, Yang H, Chen P. Organ-on-a-chip meets artificial intelligence in drug evaluation. Theranostics 2023; 13:4526-4558. [PMID: 37649608 PMCID: PMC10465229 DOI: 10.7150/thno.87266] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/02/2023] [Indexed: 09/01/2023] Open
Abstract
Drug evaluation has always been an important area of research in the pharmaceutical industry. However, animal welfare protection and other shortcomings of traditional drug development models pose obstacles and challenges to drug evaluation. Organ-on-a-chip (OoC) technology, which simulates human organs on a chip of the physiological environment and functionality, and with high fidelity reproduction organ-level of physiology or pathophysiology, exhibits great promise for innovating the drug development pipeline. Meanwhile, the advancement in artificial intelligence (AI) provides more improvements for the design and data processing of OoCs. Here, we review the current progress that has been made to generate OoC platforms, and how human single and multi-OoCs have been used in applications, including drug testing, disease modeling, and personalized medicine. Moreover, we discuss issues facing the field, such as large data processing and reproducibility, and point to the integration of OoCs and AI in data analysis and automation, which is of great benefit in future drug evaluation. Finally, we look forward to the opportunities and challenges faced by the coupling of OoCs and AI. In summary, advancements in OoCs development, and future combinations with AI, will eventually break the current state of drug evaluation.
Collapse
Affiliation(s)
- Shiwen Deng
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Caifeng Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Junxian Cao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhao Cui
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jiang Du
- Yunnan Biovalley Pharmaceutical Co., Ltd, Kunming 650503, China
| | - Zheng Fu
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Hongjun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Peng Chen
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Yunnan Biovalley Pharmaceutical Co., Ltd, Kunming 650503, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| |
Collapse
|
40
|
Ramalhete L, Vigia E, Araújo R, Marques HP. Proteomics-Driven Biomarkers in Pancreatic Cancer. Proteomes 2023; 11:24. [PMID: 37606420 PMCID: PMC10443269 DOI: 10.3390/proteomes11030024] [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: 06/30/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/23/2023] Open
Abstract
Pancreatic cancer is a devastating disease that has a grim prognosis, highlighting the need for improved screening, diagnosis, and treatment strategies. Currently, the sole biomarker for pancreatic ductal adenocarcinoma (PDAC) authorized by the U.S. Food and Drug Administration is CA 19-9, which proves to be the most beneficial in tracking treatment response rather than in early detection. In recent years, proteomics has emerged as a powerful tool for advancing our understanding of pancreatic cancer biology and identifying potential biomarkers and therapeutic targets. This review aims to offer a comprehensive survey of proteomics' current status in pancreatic cancer research, specifically accentuating its applications and its potential to drastically enhance screening, diagnosis, and treatment response. With respect to screening and diagnostic precision, proteomics carries the capacity to augment the sensitivity and specificity of extant screening and diagnostic methodologies. Nonetheless, more research is imperative for validating potential biomarkers and establishing standard procedures for sample preparation and data analysis. Furthermore, proteomics presents opportunities for unveiling new biomarkers and therapeutic targets, as well as fostering the development of personalized treatment strategies based on protein expression patterns associated with treatment response. In conclusion, proteomics holds great promise for advancing our understanding of pancreatic cancer biology and improving patient outcomes. It is essential to maintain momentum in investment and innovation in this arena to unearth more groundbreaking discoveries and transmute them into practical diagnostic and therapeutic strategies in the clinical context.
Collapse
Affiliation(s)
- Luís Ramalhete
- Blood and Transplantation Center of Lisbon—Instituto Português do Sangue e da Transplantação, Alameda das Linhas de Torres, n° 117, 1769-001 Lisbon, Portugal
- Nova Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
- iNOVA4Health—Advancing Precision Medicine, RG11: Reno-Vascular Diseases Group, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
| | - Emanuel Vigia
- Nova Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
- Centro Hospitalar de Lisboa Central, Department of Hepatobiliopancreatic and Transplantation, 1050-099 Lisbon, Portugal
| | - Rúben Araújo
- Nova Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
- CHRC—Comprehensive Health Research Centre, NOVA Medical School, 1150-199 Lisbon, Portugal
| | - Hugo Pinto Marques
- Nova Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
- Centro Hospitalar de Lisboa Central, Department of Hepatobiliopancreatic and Transplantation, 1050-099 Lisbon, Portugal
| |
Collapse
|
41
|
Kriegsmann M, Kriegsmann K, Steinbuss G, Zgorzelski C, Albrecht T, Heinrich S, Farkas S, Roth W, Dang H, Hausen A, Gaida MM. Implementation of deep learning in liver pathology optimizes diagnosis of benign lesions and adenocarcinoma metastasis. Clin Transl Med 2023; 13:e1299. [PMID: 37415390 PMCID: PMC10326372 DOI: 10.1002/ctm2.1299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 05/28/2023] [Indexed: 07/08/2023] Open
Abstract
INTRODUCTION Differentiation of histologically similar structures in the liver, including anatomical structures, benign bile duct lesions, or common types of liver metastases, can be challenging with conventional histological tissue sections alone. Accurate histopathological classification is paramount for the diagnosis and adequate treatment of the disease. Deep learning algorithms have been proposed for objective and consistent assessment of digital histopathological images. MATERIALS AND METHODS In the present study, we trained and evaluated deep learning algorithms based on the EfficientNetV2 and ResNetRS architectures to discriminate between different histopathological classes. For the required dataset, specialized surgical pathologists annotated seven different histological classes, including different non-neoplastic anatomical structures, benign bile duct lesions, and liver metastases from colorectal and pancreatic adenocarcinoma in a large patient cohort. Annotation resulted in a total of 204.159 image patches, followed by discrimination analysis using our deep learning models. Model performance was evaluated on validation and test data using confusion matrices. RESULTS Evaluation of the test set based on tiles and cases revealed overall highly satisfactory prediction capability of our algorithm for the different histological classes, resulting in a tile accuracy of 89% (38 413/43 059) and case accuracy of 94% (198/211). Importantly, the separation of metastasis versus benign lesions was certainly confident on case level, confirming the classification model performed with high diagnostic accuracy. Moreover, the whole curated raw data set is made publically available. CONCLUSIONS Deep learning is a promising approach in surgical liver pathology supporting decision making in personalized medicine.
Collapse
Affiliation(s)
- Mark Kriegsmann
- Institute of PathologyHeidelberg UniversityHeidelbergGermany
- Pathology WiesbadenWiesbadenGermany
| | - Katharina Kriegsmann
- Department of HematologyOncology and RheumatologyHeidelberg UniversityHeidelbergGermany
- Laborarztpraxis Rhein‐Main MVZ GbRFrankfurt am MainFrankfurtGermany
| | - Georg Steinbuss
- Department of HematologyOncology and RheumatologyHeidelberg UniversityHeidelbergGermany
| | | | - Thomas Albrecht
- Institute of PathologyHeidelberg UniversityHeidelbergGermany
| | - Stefan Heinrich
- Department of SurgeryJGU‐MainzUniversity Medical Center MainzMainzGermany
| | - Stefan Farkas
- Department of SurgerySt. Josefs‐ HospitalWiesbadenGermany
| | - Wilfried Roth
- Institute of PathologyJGU‐MainzUniversity Medical Center MainzMainzGermany
| | - Hien Dang
- Department of SurgeryDepartment of Surgical ResearchThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Anne Hausen
- Institute of PathologyJGU‐MainzUniversity Medical Center MainzMainzGermany
| | - Matthias M. Gaida
- Institute of PathologyJGU‐MainzUniversity Medical Center MainzMainzGermany
- TRONJGU‐MainzTranslational Oncology at the University Medical CenterMainzGermany
- Research Center for ImmunotherapyJGU‐MainzUniversity Medical Center MainzMainzGermany
| |
Collapse
|
42
|
Digiacomo L, Quagliarini E, Pozzi D, Coppola R, Caracciolo G, Caputo D. Stratifying Risk for Pancreatic Cancer by Multiplexed Blood Test. Cancers (Basel) 2023; 15:cancers15112983. [PMID: 37296945 DOI: 10.3390/cancers15112983] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/20/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease, for which mortality closely parallels incidence. So far, the available techniques for PDAC detection are either too invasive or not sensitive enough. To overcome this limitation, here we present a multiplexed point-of-care test that provides a "risk score" for each subject under investigation, by combining systemic inflammatory response biomarkers, standard laboratory tests, and the most recent nanoparticle-enabled blood (NEB) tests. The former parameters are routinely evaluated in clinical practice, whereas NEB tests have been recently proven as promising tools to assist in PDAC diagnosis. Our results revealed that PDAC patients and healthy subjects can be distinguished accurately (i.e., 88.9% specificity, 93.6% sensitivity) by the presented multiplexed point-of-care test, in a quick, non-invasive, and highly cost-efficient way. Furthermore, the test allows for the definition of a "risk threshold", which can help clinicians to trace the optimal diagnostic and therapeutic care pathway for each patient. For these reasons, we envision that this work may accelerate progress in the early detection of PDAC and contribute to the design of screening programs for high-risk populations.
Collapse
Affiliation(s)
- Luca Digiacomo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy
| | - Erica Quagliarini
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy
| | - Daniela Pozzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy
| | - Roberto Coppola
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy
- Research Unit of Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| | - Giulio Caracciolo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy
| | - Damiano Caputo
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy
- Research Unit of Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| |
Collapse
|
43
|
Faur AC, Lazar DC, Ghenciu LA. Artificial intelligence as a noninvasive tool for pancreatic cancer prediction and diagnosis. World J Gastroenterol 2023; 29:1811-1823. [PMID: 37032728 PMCID: PMC10080704 DOI: 10.3748/wjg.v29.i12.1811] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/23/2022] [Accepted: 03/15/2023] [Indexed: 03/28/2023] Open
Abstract
Pancreatic cancer (PC) has a low incidence rate but a high mortality, with patients often in the advanced stage of the disease at the time of the first diagnosis. If detected, early neoplastic lesions are ideal for surgery, offering the best prognosis. Preneoplastic lesions of the pancreas include pancreatic intraepithelial neoplasia and mucinous cystic neoplasms, with intraductal papillary mucinous neoplasms being the most commonly diagnosed. Our study focused on predicting PC by identifying early signs using noninvasive techniques and artificial intelligence (AI). A systematic English literature search was conducted on the PubMed electronic database and other sources. We obtained a total of 97 studies on the subject of pancreatic neoplasms. The final number of articles included in our study was 44, 34 of which focused on the use of AI algorithms in the early diagnosis and prediction of pancreatic lesions. AI algorithms can facilitate diagnosis by analyzing massive amounts of data in a short period of time. Correlations can be made through AI algorithms by expanding image and electronic medical records databases, which can later be used as part of a screening program for the general population. AI-based screening models should involve a combination of biomarkers and medical and imaging data from different sources. This requires large numbers of resources, collaboration between medical practitioners, and investment in medical infrastructures.
Collapse
Affiliation(s)
- Alexandra Corina Faur
- Department of Anatomy and Embriology, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Timișoara 300041, Timiș, Romania
| | - Daniela Cornelia Lazar
- Department V of Internal Medicine I, Discipline of Internal Medicine IV, University of Medicine and Pharmacy “Victor Babes” Timișoara, Timișoara 300041, Timiș, Romania
| | - Laura Andreea Ghenciu
- Department III, Discipline of Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy, Timișoara 300041, Timiș, Romania
| |
Collapse
|
44
|
Quero G, De Sio D, Fiorillo C, Menghi R, Rosa F, Massimiani G, Laterza V, Lucinato C, Galiandro F, Papa V, Salvatore L, Bensi M, Tortorelli AP, Tondolo V, Alfieri S. The role of the multidisciplinary tumor board (MDTB) in the assessment of pancreatic cancer diagnosis and resectability: A tertiary referral center experience. Front Surg 2023; 10:1119557. [PMID: 36874464 PMCID: PMC9981784 DOI: 10.3389/fsurg.2023.1119557] [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: 12/08/2022] [Accepted: 01/31/2023] [Indexed: 02/19/2023] Open
Abstract
Background The introduction of multidisciplinary tumor boards (MDTBs) for the diagnostic and therapeutic pathway of several oncological disease significantly ameliorated patients' outcomes. However, only few evidences are currently present on the potential impact of the MDTB on pancreatic cancer (PC) management. Aim of this study is to report how MDTB may influence PC diagnosis and treatment, with particular focus on PC resectability assessment and the correspondence between MDTB definition of resectability and intraoperative findings. Methods All patients with a proven or suspected diagnosis of PC discussed at the MDTB between 2018 and 2020 were included in the study. An evaluation of diagnosis, tumor response to oncological/radiation therapy and resectability before and after the MDTB was conducted. Moreover, a comparison between the MDTB resectability assessment and the intraoperative findings was performed. Results A total of 487 cases were included in the analysis: 228 (46.8%) for diagnosis evaluation, 75 (15.4%) for tumor response assessment after/during medical treatment, 184 (37.8%) for PC resectability assessment. As a whole, MDTB led to a change in treatment management in 89 cases (18.3%): 31/228 (13.6%) in the diagnosis group, 13/75 (17.3%) in the assessment of treatment response cohort and 45/184 (24.4%) in the PC resectability evaluation group. As a whole, 129 patients were given indication to surgery. Surgical resection was accomplished in 121 patients (93.7%), with a concordance rate of resectability between MDTB discussion and intraoperative findings of 91.5%. Concordance rate was 99% for resectable lesions and 64.3% for borderline PCs. Conclusions MDTB discussion consistently influences PC management, with significant variations in terms of diagnosis, tumor response assessment and resectability. In this last regard, MDTB discussion plays a key role, as demonstrated by the high concordance rate between MDTB resectability definition and intraoperative findings.
Collapse
Affiliation(s)
- Giuseppe Quero
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy.,Gemelli Pancreatic Advanced Research Center (CRMPG), Università Cattolica del Sacro Cuore di Roma, Rome, Italy
| | - Davide De Sio
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy
| | - Claudio Fiorillo
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy
| | - Roberta Menghi
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy.,Gemelli Pancreatic Advanced Research Center (CRMPG), Università Cattolica del Sacro Cuore di Roma, Rome, Italy
| | - Fausto Rosa
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy.,Gemelli Pancreatic Advanced Research Center (CRMPG), Università Cattolica del Sacro Cuore di Roma, Rome, Italy
| | - Giuseppe Massimiani
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy
| | - Vito Laterza
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy
| | - Chiara Lucinato
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy
| | - Federica Galiandro
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy
| | - Valerio Papa
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy.,Gemelli Pancreatic Advanced Research Center (CRMPG), Università Cattolica del Sacro Cuore di Roma, Rome, Italy
| | - Lisa Salvatore
- Gemelli Pancreatic Advanced Research Center (CRMPG), Università Cattolica del Sacro Cuore di Roma, Rome, Italy.,Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Maria Bensi
- Gemelli Pancreatic Advanced Research Center (CRMPG), Università Cattolica del Sacro Cuore di Roma, Rome, Italy.,Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Antonio Pio Tortorelli
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy
| | - Vincenzo Tondolo
- General Surgery Unit, Fatebenefratelli Isola Tiberina - Gemelli Isola, Via di Ponte Quattro Capi, Roma, Italy
| | - Sergio Alfieri
- Pancreatic Surgery Unit, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli, Rome, Italy.,Gemelli Pancreatic Advanced Research Center (CRMPG), Università Cattolica del Sacro Cuore di Roma, Rome, Italy
| |
Collapse
|
45
|
Dahiya DS, Al-Haddad M, Chandan S, Gangwani MK, Aziz M, Mohan BP, Ramai D, Canakis A, Bapaye J, Sharma N. Artificial Intelligence in Endoscopic Ultrasound for Pancreatic Cancer: Where Are We Now and What Does the Future Entail? J Clin Med 2022; 11:jcm11247476. [PMID: 36556092 PMCID: PMC9786876 DOI: 10.3390/jcm11247476] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Pancreatic cancer is a highly lethal disease associated with significant morbidity and mortality. In the United States (US), the overall 5-year relative survival rate for pancreatic cancer during the 2012-2018 period was 11.5%. However, the cancer stage at diagnosis strongly influences relative survival in these patients. Per the National Cancer Institute (NCI) statistics for 2012-2018, the 5-year relative survival rate for patients with localized disease was 43.9%, while it was 3.1% for patients with distant metastasis. The poor survival rates are primarily due to the late development of clinical signs and symptoms. Hence, early diagnosis is critical in improving treatment outcomes. In recent years, artificial intelligence (AI) has gained immense popularity in gastroenterology. AI-assisted endoscopic ultrasound (EUS) models have been touted as a breakthrough in the early detection of pancreatic cancer. These models may also accurately differentiate pancreatic cancer from chronic pancreatitis and autoimmune pancreatitis, which mimics pancreatic cancer on radiological imaging. In this review, we detail the application of AI-assisted EUS models for pancreatic cancer detection. We also highlight the utility of AI-assisted EUS models in differentiating pancreatic cancer from radiological mimickers. Furthermore, we discuss the current limitations and future applications of AI technology in EUS for pancreatic cancers.
Collapse
Affiliation(s)
- Dushyant Singh Dahiya
- Department of Internal Medicine, Central Michigan University College of Medicine, Saginaw, MI 48601, USA
- Correspondence: ; Tel.: +1-(678)-602-1176
| | - Mohammad Al-Haddad
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Saurabh Chandan
- Division of Gastroenterology and Hepatology, CHI Creighton University Medical Center, Omaha, NE 68131, USA
| | - Manesh Kumar Gangwani
- Department of Internal Medicine, The University of Toledo Medical Center, Toledo, OH 43614, USA
| | - Muhammad Aziz
- Department of Gastroenterology, The University of Toledo Medical Center, Toledo, OH 43614, USA
| | - Babu P. Mohan
- Division of Gastroenterology and Hepatology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Daryl Ramai
- Division of Gastroenterology and Hepatology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Andrew Canakis
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jay Bapaye
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY 14621, USA
| | - Neil Sharma
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Parkview Cancer Institute, Fort Wayne, IN 46845, USA
- Interventional Oncology & Surgical Endoscopy Programs (IOSE), Parkview Health, Fort Wayne, IN 46845, USA
| |
Collapse
|