1
|
Xia C, Zuo M, Lin Z, Deng L, Rao Y, Chen W, Chen J, Yao W, Hu M. Multimodal Deep Learning Fusing Clinical and Radiomics Scores for Prediction of Early-Stage Lung Adenocarcinoma Lymph Node Metastasis. Acad Radiol 2025; 32:2977-2989. [PMID: 39730249 DOI: 10.1016/j.acra.2024.12.018] [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: 09/30/2024] [Revised: 11/27/2024] [Accepted: 12/09/2024] [Indexed: 12/29/2024]
Abstract
RATIONALE AND OBJECTIVES To develop and validate a multimodal deep learning (DL) model based on computed tomography (CT) images and clinical knowledge to predict lymph node metastasis (LNM) in early lung adenocarcinoma. MATERIALS AND METHODS A total of 724 pathologically confirmed early invasive lung adenocarcinoma patients were retrospectively included from two centers. Clinical and CT semantic features of the patients were collected, and 3D radiomics features were extracted from nonenhanced CT images. We proposed a multimodal feature fusion DL network based on the InceptionResNetV2 architecture, which can effectively extract and integrate image and clinical knowledge to predict LNM. RESULTS A total of 524 lung adenocarcinoma patients from Center 1 were randomly divided into training (n=418) and internal validation (n=106) sets in a 4:1 ratio, while 200 lung adenocarcinoma patients from Center 2 served as the independent test set. Among the 16 collected clinical and imaging features, 8 were selected: gender, serum carcinoembryonic antigen, cytokeratin 19 fragment antigen 21-1, neuron-specific enolase, tumor size, location, density, and centrality. From the 1595 extracted radiomics features, six key features were identified. The CS-RS-DL fusion model achieved the highest area under the receiver operating characteristic curve in both the internal validation set (0.877) and the independent test set (0.906) compared to other models. The Delong test results for the independent test set indicated that the CS-RS-DL model significantly outperformed the clinical model (0.844), radiomics model (0.850), CS-RS model (0.872), single DL model (0.848), and the CS-DL model (0.875) (all P<0.05). Additionally, the CS-RS-DL model exhibited the highest sensitivity (0.941) and average precision (0.642). CONCLUSION The knowledge derived from clinical, radiomics, and DL is complementary in predicting LNM in lung adenocarcinoma. The integration of clinical and radiomics scores through DL can significantly improve the accuracy of lymph node status assessment.
Collapse
Affiliation(s)
- Chengcheng Xia
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330006, China (C.X., L.D., W.C., M.H.); Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang 330006, China (C.X., L.D., W.C., M.H.)
| | - Minjing Zuo
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China (M.Z.); Intelligent Medical Imaging of Jiangxi Key Laboratory, Nanchang 330006, China (M.Z.)
| | - Ze Lin
- Department of Radiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430022, China (Z.L.); Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan 430022, China (Z.L.)
| | - Libin Deng
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330006, China (C.X., L.D., W.C., M.H.); Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang 330006, China (C.X., L.D., W.C., M.H.)
| | - Yulian Rao
- Wanli District Center for Disease Control and Prevention of Nanchang, Nanchang 330004, China (Y.R.)
| | - Wenxiang Chen
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330006, China (C.X., L.D., W.C., M.H.); Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang 330006, China (C.X., L.D., W.C., M.H.)
| | - Jinqin Chen
- Jiangxi Medical College, Nanchang University, Nanchang, China (J.C.)
| | - Weirong Yao
- Department of Oncology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China (W.Y.)
| | - Min Hu
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330006, China (C.X., L.D., W.C., M.H.); Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang 330006, China (C.X., L.D., W.C., M.H.).
| |
Collapse
|
2
|
Hosseini H, Heydari S, Hushmandi K, Daneshi S, Raesi R. Bone tumors: a systematic review of prevalence, risk determinants, and survival patterns. BMC Cancer 2025; 25:321. [PMID: 39984867 PMCID: PMC11846205 DOI: 10.1186/s12885-025-13720-0] [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: 01/19/2025] [Accepted: 02/12/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Though relatively rare, bone tumors significantly impact patient health and treatment outcomes. OBJECTIVE This systematic review analyzes the incidence, types, survival rates, and risk factors associated with bone tumors, including both benign and malignant forms. METHODS This systematic review was conducted using the keywords "bone tumors," "epidemiology," "benign bone tumors," "malignant bone tumors," "osteosarcoma," "Ewing sarcoma," "chondrosarcoma," "risk factors," and "survival" in electronic databases including PubMed, Scopus, Web of Science, and Google Scholar from 2000 to 2024. The search strategy was based on the PRISMA statement. Finally, 9 articles were selected for inclusion in the study. RESULTS The systematic review highlights that primary bone tumors can be classified into benign and malignant types, with osteosarcoma being the most prevalent malignant form, particularly among adolescents and young adults. The epidemiology of bone tumors is influenced by factors such as age, gender, geographic location, and genetic predispositions. Recent advancements in imaging techniques have improved the detection of these tumors, contributing to an increasing recognition of their prevalence. Data shows that the limited-duration prevalence of malignant bone tumors has increased significantly. This increase is from 0.00069% in 2000 to 0.00749% in 2018, indicating an increasing recognition and diagnosis of these rare tumors over time. Survival rates vary significantly by tumor type, with approximately 50-60% for osteosarcoma and around 70% for Ewing's sarcoma, though these rates decrease with metastasis. Key risk factors identified include genetic predispositions such as Li-Fraumeni syndrome and TP53 mutations, environmental exposures like radiation, and growth patterns related to height. CONCLUSION The review highlights the importance of early diagnosis and treatment intervention, as survival rates are significantly better for patients with localized disease compared to those with metastatic conditions. The observed variations in survival rates across different tumor types underscore the need for tailored treatment strategies. Key risk factors include genetic predispositions and environmental exposures, highlighting the need for targeted screening and ongoing research to enhance diagnostic accuracy and treatment strategies.
Collapse
Affiliation(s)
- Hasan Hosseini
- Department of Orthopedics, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sina Heydari
- School of Medicine, Imam Khomeini Hospital, Jiroft University of Medical Science, Jiroft, Iran
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Rasoul Raesi
- Department of Public Health, School of Health, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran.
- Department of Health Services Management, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Zhang J, Ding F, Guo Y, Wei X, Jing J, Xu F, Chen H, Guo Z, You Z, Liang B, Chen M, Jiang D, Niu X, Wang X, Xue Y. AI-based prediction of androgen receptor expression and its prognostic significance in prostate cancer. Sci Rep 2025; 15:3985. [PMID: 39893198 PMCID: PMC11787347 DOI: 10.1038/s41598-025-88199-7] [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: 10/17/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025] Open
Abstract
Biochemical recurrence (BCR) of prostate cancer (PCa) negatively impacts patients' post-surgery quality of life, and the traditional predictive models have shown limited accuracy. This study develops an AI-based prognostic model using deep learning that incorporates androgen receptor (AR) regional features from whole-slide images (WSIs). Data from 545 patients across two centres are used for training and validation. The model showed strong performances, with high accuracy in identifying regions with high AR expression and BCR prediction. This AI model may help identify high-risk patients, aiding in better treatment strategies, particularly in underdeveloped areas.
Collapse
Affiliation(s)
- Jiawei Zhang
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
- Department of Medical College, Southeast University, Nanjing, China
| | - Feng Ding
- Nanjing University of Information Science and Technology, Nanjing, China
| | - Yitian Guo
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
- Department of Medical College, Southeast University, Nanjing, China
| | - Xiaoying Wei
- Department of Medical College, Southeast University, Nanjing, China
- Department of Pathology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jibo Jing
- Department of Urology, Peking Union Medical College Hospital, Beijing, China
| | - Feng Xu
- Jinhu County People's Hospital, Huai'an, China
| | - Huixing Chen
- Shanghai General Hospital, Urologic Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongying Guo
- Department of Pathology, Huaian First People's Hospital, Huai'an, China
| | - Zonghao You
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
- Department of Medical College, Southeast University, Nanjing, China
| | - Baotai Liang
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
- Department of Medical College, Southeast University, Nanjing, China
| | - Ming Chen
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
- Department of Medical College, Southeast University, Nanjing, China
| | - Dongfang Jiang
- Department of Urology, The People's Hospital of Danyang, Danyang, China.
| | - Xiaobing Niu
- Department of Urology, Huaian First People's Hospital, Huai'an, China.
| | - Xiangxue Wang
- Nanjing University of Information Science and Technology, Nanjing, China.
| | - Yifeng Xue
- The Affiliated Jintan Hospital of Jiangsu University, Changzhou, China.
- Changzhou jintan first people's hospital, Changzhou, China.
| |
Collapse
|
4
|
Rai HM, Yoo J, Dashkevych S. Transformative Advances in AI for Precise Cancer Detection: A Comprehensive Review of Non-Invasive Techniques. ARCHIVES OF COMPUTATIONAL METHODS IN ENGINEERING 2025. [DOI: 10.1007/s11831-024-10219-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 12/07/2024] [Indexed: 03/02/2025]
|
5
|
Huang T, Huang X, Yin H. Deep learning methods for improving the accuracy and efficiency of pathological image analysis. Sci Prog 2025; 108:368504241306830. [PMID: 39814425 PMCID: PMC11736776 DOI: 10.1177/00368504241306830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
This study presents a novel integration of two advanced deep learning models, U-Net and EfficientNetV2, to achieve high-precision segmentation and rapid classification of pathological images. A key innovation is the development of a new heatmap generation algorithm, which leverages meticulous image preprocessing, data enhancement strategies, ensemble learning, attention mechanisms, and deep feature fusion techniques. This algorithm not only produces highly accurate and interpretatively rich heatmaps but also significantly improves the accuracy and efficiency of pathological image analysis. Unlike existing methods, our approach integrates these advanced techniques into a cohesive framework, enhancing its ability to reveal critical features in pathological images. Rigorous experimental validation demonstrated that our algorithm excels in key performance indicators such as accuracy, recall rate, and processing speed, underscoring its potential for broader applications in pathological image analysis and beyond.
Collapse
Affiliation(s)
- Tangsen Huang
- School of Communication Engineering, Hangzhou Dianzi University, Hangzhou, China
- School of Mathematics and Computer Science, Lishui University, Lishui, China
- School of Information Engineering, Hunan University of Science and Engineering, Yongzhou, China
| | - Xingru Huang
- School of Communication Engineering, Hangzhou Dianzi University, Hangzhou, China
| | - Haibing Yin
- School of Communication Engineering, Hangzhou Dianzi University, Hangzhou, China
- School of Mathematics and Computer Science, Lishui University, Lishui, China
| |
Collapse
|
6
|
Shahzadi M, Rafique H, Waheed A, Naz H, Waheed A, Zokirova FR, Khan H. Artificial intelligence for chimeric antigen receptor-based therapies: a comprehensive review of current applications and future perspectives. Ther Adv Vaccines Immunother 2024; 12:25151355241305856. [PMID: 39691280 PMCID: PMC11650588 DOI: 10.1177/25151355241305856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Using artificial intelligence (AI) to enhance chimeric antigen receptor (CAR)-based therapies' design, production, and delivery is a novel and promising approach. This review provides an overview of the current applications and challenges of AI for CAR-based therapies and suggests some directions for future research and development. This paper examines some of the recent advances of AI for CAR-based therapies, for example, using deep learning (DL) to design CARs that target multiple antigens and avoid antigen escape; using natural language processing to extract relevant information from clinical reports and literature; using computer vision to analyze the morphology and phenotype of CAR cells; using reinforcement learning to optimize the dose and schedule of CAR infusion; and using AI to predict the efficacy and toxicity of CAR-based therapies. These applications demonstrate the potential of AI to improve the quality and efficiency of CAR-based therapies and to provide personalized and precise treatments for cancer patients. However, there are also some challenges and limitations of using AI for CAR-based therapies, for example, the lack of high-quality and standardized data; the need for validation and verification of AI models; the risk of bias and error in AI outputs; the ethical, legal, and social issues of using AI for health care; and the possible impact of AI on the human role and responsibility in cancer immunotherapy. It is important to establish a multidisciplinary collaboration among researchers, clinicians, regulators, and patients to address these challenges and to ensure the safe and responsible use of AI for CAR-based therapies.
Collapse
Affiliation(s)
- Muqadas Shahzadi
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Hamad Rafique
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Ahmad Waheed
- Department of Zoology, Faculty of Life Sciences, University of Okara, 2 KM Lahore Road, Renala Khurd, Okara 56130, Punjab, Pakistan
| | - Hina Naz
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Atifa Waheed
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | | | - Humera Khan
- Department of Biochemistry, Sahiwal Medical College, Sahiwal, Pakistan
| |
Collapse
|
7
|
Rai HM, Yoo J, Razaque A. Comparative analysis of machine learning and deep learning models for improved cancer detection: A comprehensive review of recent advancements in diagnostic techniques. EXPERT SYSTEMS WITH APPLICATIONS 2024; 255:124838. [DOI: 10.1016/j.eswa.2024.124838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
|
8
|
Wang F, Song Y, Xu H, Liu J, Tang F, Yang D, Yang D, Liang W, Ren L, Wang J, Luo X, Zhou Y, Zeng X, Dan H, Chen Q. Prediction of the short-term efficacy and recurrence of photodynamic therapy in the treatment of oral leukoplakia based on deep learning. Photodiagnosis Photodyn Ther 2024; 48:104236. [PMID: 38851310 DOI: 10.1016/j.pdpdt.2024.104236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND The treatment of oral leukoplakia (OLK) with aminolaevulinic acid photodynamic therapy (ALA-PDT) is widespread. Nonetheless, there is variation in efficacy. Therefore, this study constructed a model for predicting the short-term efficacy and recurrence of OLK after ALA-PDT. METHODS The short-term efficacy and recurrence of ALA-PDT were calculated by statistical analysis, and the relevant influencing factors were analyzed by Logistic regression and COX regression model. Finally, prediction models for total response (TR) rate, complete response (CR) rate and recurrence in OLK patients after ALA-PDT treatment were established. Features from pathology sections were extracted using deep learning autoencoder and combined with clinical variables to improve prediction performance of the model. RESULTS The logistic regression analysis showed that the non-homogeneous (OR: 4.911, P: 0.023) OLK and lesions with moderate to severe epithelial dysplasia (OR: 4.288, P: 0.042) had better short-term efficacy. The area under receiver operating characteristic curve (AUC) of CR, TR and recurrence predict models after the ALA-PDT treatment of OLK patients is 0.872, 0.718, and 0.564, respectively. Feature extraction revealed an association between inflammatory cell infiltration in the lamina propria and recurrence after PDT. Combining clinical variables and deep learning improved the performance of recurrence model by more than 30 %. CONCLUSIONS ALA-PDT has excellent short-term efficacy in the management of OLK but the recurrence rate was high. Prediction model based on clinicopathological characteristics has excellent predictive effect for short-term efficacy but limited effect for recurrence. The use of deep learning and pathology images greatly improves predictive value of the models.
Collapse
Affiliation(s)
- Fei Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yansong Song
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Hao Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jiaxin Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Fan Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China; Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Cancer Center of Zhejiang University, Hangzhou 310000, PR China
| | - Dan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Dan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wenhui Liang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ling Ren
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jiongke Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xiaobo Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yu Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Hongxia Dan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China; Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Cancer Center of Zhejiang University, Hangzhou 310000, PR China.
| |
Collapse
|
9
|
Al-Ostoot FH, Salah S, Khanum SA. An Overview of Cancer Biology, Pathophysiological Development and It's Treatment Modalities: Current Challenges of Cancer anti-Angiogenic Therapy. Cancer Invest 2024; 42:559-604. [PMID: 38874308 DOI: 10.1080/07357907.2024.2361295] [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: 02/17/2021] [Revised: 11/22/2021] [Accepted: 05/25/2024] [Indexed: 06/15/2024]
Abstract
A number of conditions and factors can cause the transformation of normal cells in the body into malignant tissue by changing the normal functions of a wide range of regulatory, apoptotic, and signal transduction pathways. Despite the current deficiency in fully understanding the mechanism of cancer action accurately and clearly, numerous genes and proteins that are causally involved in the initiation, progression, and metastasis of cancer have been identified. But due to the lack of space and the abundance of details on this complex topic, we have emphasized here more recent advances in our understanding of the principles implied tumor cell transformation, development, invasion, angiogenesis, and metastasis. Inhibition of angiogenesis is a significant strategy for the treatment of various solid tumors, that essentially depend on cutting or at least limiting the supply of blood to micro-regions of tumors, leading to pan-hypoxia and pan-necrosis inside solid tumor tissues. Researchers have continued to enhance the efficiency of anti-angiogenic drugs over the past two decades, to identify their potential in the drug interaction, and to discover reasonable interpretations for possible resistance to treatment. In this review, we have discussed an overview of cancer history and recent methods use in cancer therapy, focusing on anti-angiogenic inhibitors targeting angiogenesis formation. Further, this review has explained the molecular mechanism of action of these anti-angiogenic inhibitors in various tumor types and their limitations use. In addition, we described the synergistic mechanisms of immunotherapy and anti-angiogenic therapy and summarizes current clinical trials of these combinations. Many phase III trials found that combining immunotherapy and anti-angiogenic therapy improved survival. Therefore, targeting the source supply of cancer cells to grow and spread with new anti-angiogenic agents in combination with different conventional therapy is a novel method to reduce cancer progression. The aim of this paper is to overview the varying concepts of cancer focusing on mechanisms involved in tumor angiogenesis and provide an overview of the recent trends in anti-angiogenic strategies for cancer therapy.
Collapse
Affiliation(s)
- Fares Hezam Al-Ostoot
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysuru, India
- Department of Biochemistry, Faculty of Education & Science, Albaydha University, Al-Baydha, Yemen
| | - Salma Salah
- Faculty of Medicine and Health Sciences, Thamar University, Dhamar, Yemen
| | - Shaukath Ara Khanum
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysuru, India
| |
Collapse
|
10
|
Farjami E, Mahjoob M. Multiscale modeling of the dynamic growth of cancerous tumors under the influence of chemotherapy drugs. Comput Methods Biomech Biomed Engin 2024; 27:919-930. [PMID: 37227061 DOI: 10.1080/10255842.2023.2215368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 05/12/2023] [Indexed: 05/26/2023]
Abstract
A comprehensive model of chemotherapy treatment of cancer can help us to optimize the drug administration/dosage and improve the treatment outcome. In the present study, a multiscale mathematical model of tumor growth during chemotherapy treatment is developed to predict its response to the medication and cancer progression. The modeling is a continuous multiscale simulation consisting of three tissue phases including cancer cells, normal cells, and extracellular matrix. In addition to the drug administration, the impacts of immune cells, programmed cell death, nutrient competition, and glucose concentration are included. The outputs of our mathematical model conform to the published experimental and clinical data, and it can be used in optimizing chemotherapy, and personalized cancer treatment.
Collapse
Affiliation(s)
- Emad Farjami
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mohammad Mahjoob
- Department of Engineering, School of Engineering, Science and Technology, CCSU, New Britain, CT, USA
| |
Collapse
|
11
|
Rich K, Tosefsky K, Martin KC, Bashashati A, Yip S. Practical Application of Deep Learning in Diagnostic Neuropathology-Reimagining a Histological Asset in the Era of Precision Medicine. Cancers (Basel) 2024; 16:1976. [PMID: 38893099 PMCID: PMC11171052 DOI: 10.3390/cancers16111976] [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: 04/07/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
In the past few decades, neuropathology has experienced several paradigm shifts with the introduction of new technologies. Deep learning, a rapidly progressing subfield of machine learning, seems to be the next innovation to alter the diagnostic workflow. In this review, we will explore the recent changes in the field of neuropathology and how this has led to an increased focus on molecular features in diagnosis and prognosis. Then, we will examine the work carried out to train deep learning models for various diagnostic tasks in neuropathology, as well as the machine learning frameworks they used. Focus will be given to both the challenges and successes highlighted therein, as well as what these trends may tell us about future roadblocks in the widespread adoption of this new technology. Finally, we will touch on recent trends in deep learning, as applied to digital pathology more generally, and what this may tell us about the future of deep learning applications in neuropathology.
Collapse
Affiliation(s)
- Katherine Rich
- Bioinformatics Graduate Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
| | - Kira Tosefsky
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (K.T.); (K.C.M.)
| | - Karina C. Martin
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (K.T.); (K.C.M.)
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Ali Bashashati
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Stephen Yip
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (K.T.); (K.C.M.)
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
12
|
Mou X, Wang P, Sun J, Chen X, Du L, Zhan Q, Xia J, Yang T, Fang Z. A Novel Approach for the Detection and Severity Grading of Chronic Obstructive Pulmonary Disease Based on Transformed Volumetric Capnography. Bioengineering (Basel) 2024; 11:530. [PMID: 38927766 PMCID: PMC11200784 DOI: 10.3390/bioengineering11060530] [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: 04/15/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD), as the third leading cause of death worldwide, is a major global health issue. The early detection and grading of COPD are pivotal for effective treatment. Traditional spirometry tests, requiring considerable physical effort and strict adherence to quality standards, pose challenges in COPD diagnosis. Volumetric capnography (VCap), which can be performed during natural breathing without requiring additional compliance, presents a promising alternative tool. In this study, the dataset comprised 279 subjects with normal pulmonary function and 148 patients diagnosed with COPD. We introduced a novel quantitative analysis method for VCap. Volumetric capnograms were converted into two-dimensional grayscale images through the application of Gramian Angular Field (GAF) transformation. Subsequently, a multi-scale convolutional neural network, CapnoNet, was conducted to extract features and facilitate classification. To improve CapnoNet's performance, two data augmentation techniques were implemented. The proposed model exhibited a detection accuracy for COPD of 95.83%, with precision, recall, and F1 measures of 95.21%, 95.70%, and 95.45%, respectively. In the task of grading the severity of COPD, the model attained an accuracy of 96.36%, complemented by precision, recall, and F1 scores of 88.49%, 89.99%, and 89.15%, respectively. This work provides a new perspective for the quantitative analysis of volumetric capnography and demonstrates the strong performance of the proposed CapnoNet in the diagnosis and grading of COPD. It offers direction and an effective solution for the clinical application of capnography.
Collapse
Affiliation(s)
- Xiuying Mou
- Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100094, China; (X.M.); (P.W.); (J.S.); (X.C.); (L.D.)
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng Wang
- Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100094, China; (X.M.); (P.W.); (J.S.); (X.C.); (L.D.)
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Sun
- Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100094, China; (X.M.); (P.W.); (J.S.); (X.C.); (L.D.)
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianxiang Chen
- Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100094, China; (X.M.); (P.W.); (J.S.); (X.C.); (L.D.)
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lidong Du
- Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100094, China; (X.M.); (P.W.); (J.S.); (X.C.); (L.D.)
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing 100029, China; (Q.Z.); (J.X.)
| | - Jingen Xia
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing 100029, China; (Q.Z.); (J.X.)
| | - Ting Yang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing 100029, China; (Q.Z.); (J.X.)
| | - Zhen Fang
- Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100094, China; (X.M.); (P.W.); (J.S.); (X.C.); (L.D.)
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- Research Unit of Personalized Management of Chronic Respiratory Disease, Chinese Academy of Medical Sciences, Beijing 100190, China
| |
Collapse
|
13
|
Chen J, Xue Y, Ren L, Lv K, Du P, Cheng H, Sun S, Hua L, Xie Q, Wu R, Gong Y. Predicting meningioma grades and pathologic marker expression via deep learning. Eur Radiol 2024; 34:2997-3008. [PMID: 37853176 DOI: 10.1007/s00330-023-10258-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 07/05/2023] [Accepted: 07/15/2023] [Indexed: 10/20/2023]
Abstract
OBJECTIVES To establish a deep learning (DL) model for predicting tumor grades and expression of pathologic markers of meningioma. METHODS A total of 1192 meningioma patients from two centers who underwent surgical resection between September 2018 and December 2021 were retrospectively included. The pathological data and post-contrast T1-weight images for each patient were collected. The patients from institute I were subdivided into training, validation, and testing sets, while the patients from institute II served as the external testing cohort. The fine-tuned ResNet50 model based on transfer learning was adopted to classify WHO grade in the whole cohort and predict Ki-67 index, H3K27me3, and progesterone receptor (PR) status of grade 1 meningiomas. The predictive performance was evaluated by the accuracy and loss curve, confusion matrix, receiver operating characteristic curve (ROC), and area under curve (AUC). RESULTS The DL prediction model for each label achieved high predictive performance in two cohorts. For WHO grade prediction, the area under the curve (AUC) was 0.966 (95%CI 0.957-0.975) in the internal testing set and 0.669 (95%CI 0.643-0.695) in the external validation cohort. The AUC in predicting Ki-67 index, H3K27me3, and PR status were 0.905 (95%CI 0.895-0.915), 0.773 (95%CI 0.760-0.786), and 0.771 (95%CI 0.750-0.792) in the internal testing set and 0.591 (95%CI 0.562-0.620), 0.658 (95%CI 0.648-0.668), and 0.703 (95%CI 0.674-0.732) in the external validation cohort, respectively. CONCLUSION DL models can preoperatively predict meningioma grades and pathologic marker expression with favorable predictive performance. CLINICAL RELEVANCE STATEMENT Our DL model could predict meningioma grades and expression of pathologic markers and identify high-risk patients with WHO grade 1 meningioma, which would suggest a more aggressive operative intervention preoperatively and a more frequent follow-up schedule postoperatively. KEY POINTS WHO grades and some pathologic markers of meningioma were associated with therapeutic strategies and clinical outcomes. A deep learning-based approach was employed to develop a model for predicting meningioma grades and the expression of pathologic markers. Preoperative prediction of meningioma grades and the expression of pathologic markers was beneficial for clinical decision-making.
Collapse
Affiliation(s)
- Jiawei Chen
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yanping Xue
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Leihao Ren
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Kun Lv
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Peng Du
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Haixia Cheng
- Department of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuchen Sun
- Department of Neurosurgery, Shanghai International Hospital, Shanghai, China
- Department of Neurosurgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingyang Hua
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qing Xie
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China.
| | - Ruiqi Wu
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China.
| | - Ye Gong
- Department of Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institutes of Brain Science, Fudan University, Shanghai, China.
- Department of Critical Care Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Fechter T, Sachpazidis I, Baltas D. The use of deep learning in interventional radiotherapy (brachytherapy): A review with a focus on open source and open data. Z Med Phys 2024; 34:180-196. [PMID: 36376203 PMCID: PMC11156786 DOI: 10.1016/j.zemedi.2022.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022]
Abstract
Deep learning advanced to one of the most important technologies in almost all medical fields. Especially in areas, related to medical imaging it plays a big role. However, in interventional radiotherapy (brachytherapy) deep learning is still in an early phase. In this review, first, we investigated and scrutinised the role of deep learning in all processes of interventional radiotherapy and directly related fields. Additionally, we summarised the most recent developments. For better understanding, we provide explanations of key terms and approaches to solving common deep learning problems. To reproduce results of deep learning algorithms both source code and training data must be available. Therefore, a second focus of this work is on the analysis of the availability of open source, open data and open models. In our analysis, we were able to show that deep learning plays already a major role in some areas of interventional radiotherapy, but is still hardly present in others. Nevertheless, its impact is increasing with the years, partly self-propelled but also influenced by closely related fields. Open source, data and models are growing in number but are still scarce and unevenly distributed among different research groups. The reluctance in publishing code, data and models limits reproducibility and restricts evaluation to mono-institutional datasets. The conclusion of our analysis is that deep learning can positively change the workflow of interventional radiotherapy but there is still room for improvements when it comes to reproducible results and standardised evaluation methods.
Collapse
Affiliation(s)
- Tobias Fechter
- Division of Medical Physics, Department of Radiation Oncology, Medical Center University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Germany.
| | - Ilias Sachpazidis
- Division of Medical Physics, Department of Radiation Oncology, Medical Center University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Germany
| | - Dimos Baltas
- Division of Medical Physics, Department of Radiation Oncology, Medical Center University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Germany
| |
Collapse
|
15
|
Abid R, Hussein AA, Guru KA. Artificial Intelligence in Urology: Current Status and Future Perspectives. Urol Clin North Am 2024; 51:117-130. [PMID: 37945097 DOI: 10.1016/j.ucl.2023.06.005] [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: 11/12/2023]
Abstract
Surgical fields, especially urology, have shifted increasingly toward the use of artificial intelligence (AI). Advancements in AI have created massive improvements in diagnostics, outcome predictions, and robotic surgery. For robotic surgery to progress from assisting surgeons to eventually reaching autonomous procedures, there must be advancements in machine learning, natural language processing, and computer vision. Moreover, barriers such as data availability, interpretability of autonomous decision-making, Internet connection and security, and ethical concerns must be overcome.
Collapse
Affiliation(s)
- Rayyan Abid
- Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Ahmed A Hussein
- Department of Urology, Roswell Park Comprehensive Cancer Center
| | - Khurshid A Guru
- Department of Urology, Roswell Park Comprehensive Cancer Center.
| |
Collapse
|
16
|
Gao Y, Wang W, Yang Y, Xu Z, Lin Y, Lang T, Lei S, Xiao Y, Yang W, Huang W, Li Y. An integrated model incorporating deep learning, hand-crafted radiomics and clinical and US features to diagnose central lymph node metastasis in patients with papillary thyroid cancer. BMC Cancer 2024; 24:69. [PMID: 38216936 PMCID: PMC10787418 DOI: 10.1186/s12885-024-11838-1] [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: 07/30/2023] [Accepted: 01/03/2024] [Indexed: 01/14/2024] Open
Abstract
OBJECTIVE To evaluate the value of an integrated model incorporating deep learning (DL), hand-crafted radiomics and clinical and US imaging features for diagnosing central lymph node metastasis (CLNM) in patients with papillary thyroid cancer (PTC). METHODS This retrospective study reviewed 613 patients with clinicopathologically confirmed PTC from two institutions. The DL model and hand-crafted radiomics model were developed using primary lesion images and then integrated with clinical and US features selected by multivariate analysis to generate an integrated model. The performance was compared with junior and senior radiologists on the independent test set. SHapley Additive exPlanations (SHAP) plot and Gradient-weighted Class Activation Mapping (Grad-CAM) were used for the visualized explanation of the model. RESULTS The integrated model yielded the best performance with an AUC of 0.841. surpassing that of the hand-crafted radiomics model (0.706, p < 0.001) and the DL model (0.819, p = 0.26). Compared to junior and senior radiologists, the integrated model reduced the missed CLNM rate from 57.89% and 44.74-27.63%, and decreased the rate of unnecessary central lymph node dissection (CLND) from 29.87% and 27.27-18.18%, respectively. SHAP analysis revealed that the DL features played a primary role in the diagnosis of CLNM, while clinical and US features (such as extrathyroidal extension, tumour size, age, gender, and multifocality) provided additional support. Grad-CAM indicated that the model exhibited a stronger focus on thyroid capsule in patients with CLNM. CONCLUSION Integrated model can effectively decrease the incidence of missed CLNM and unnecessary CLND. The application of the integrated model can help improve the acceptance of AI-assisted US diagnosis among radiologists.
Collapse
Affiliation(s)
- Yang Gao
- Department of Ultrasound, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong Province, P. R. China
| | - Weizhen Wang
- Department of Ultrasound, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong Province, P. R. China
| | - Yuan Yang
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong Province, P. R. China
| | - Ziting Xu
- Department of Ultrasound, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong Province, P. R. China
| | - Yue Lin
- Department of Ultrasound, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong Province, P. R. China
| | - Ting Lang
- Department of Ultrasound, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong Province, P. R. China
| | - Shangtong Lei
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Yisheng Xiao
- Department of Ultrasound, the First People's Hospital of Foshan, Lingnan Avenue North No.81, Foshan, Guangdong Province, P. R. China
| | - Wei Yang
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong Province, P. R. China.
| | - Weijun Huang
- Department of Ultrasound, the First People's Hospital of Foshan, Lingnan Avenue North No.81, Foshan, Guangdong Province, P. R. China.
| | - Yingjia Li
- Department of Ultrasound, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong Province, P. R. China.
| |
Collapse
|
17
|
Sharma V, Singh A, Chauhan S, Sharma PK, Chaudhary S, Sharma A, Porwal O, Fuloria NK. Role of Artificial Intelligence in Drug Discovery and Target Identification in Cancer. Curr Drug Deliv 2024; 21:870-886. [PMID: 37670704 DOI: 10.2174/1567201821666230905090621] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/08/2023] [Accepted: 03/24/2023] [Indexed: 09/07/2023]
Abstract
Drug discovery and development (DDD) is a highly complex process that necessitates precise monitoring and extensive data analysis at each stage. Furthermore, the DDD process is both timeconsuming and costly. To tackle these concerns, artificial intelligence (AI) technology can be used, which facilitates rapid and precise analysis of extensive datasets within a limited timeframe. The pathophysiology of cancer disease is complicated and requires extensive research for novel drug discovery and development. The first stage in the process of drug discovery and development involves identifying targets. Cell structure and molecular functioning are complex due to the vast number of molecules that function constantly, performing various roles. Furthermore, scientists are continually discovering novel cellular mechanisms and molecules, expanding the range of potential targets. Accurately identifying the correct target is a crucial step in the preparation of a treatment strategy. Various forms of AI, such as machine learning, neural-based learning, deep learning, and network-based learning, are currently being utilised in applications, online services, and databases. These technologies facilitate the identification and validation of targets, ultimately contributing to the success of projects. This review focuses on the different types and subcategories of AI databases utilised in the field of drug discovery and target identification for cancer.
Collapse
Affiliation(s)
- Vishal Sharma
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Amit Singh
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Sanjana Chauhan
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Pramod Kumar Sharma
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Shubham Chaudhary
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Astha Sharma
- Department of Pharmacy, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| | - Omji Porwal
- Department of Pharmacognosy, Faculty of Pharmacy, Tishk International University, Erbil 44001, Iraq
| | | |
Collapse
|
18
|
Rai HM, Yoo J. Analysis of Colorectal and Gastric Cancer Classification: A Mathematical Insight Utilizing Traditional Machine Learning Classifiers. MATHEMATICS 2023; 11:4937. [DOI: 10.3390/math11244937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Cancer remains a formidable global health challenge, claiming millions of lives annually. Timely and accurate cancer diagnosis is imperative. While numerous reviews have explored cancer classification using machine learning and deep learning techniques, scant literature focuses on traditional ML methods. In this manuscript, we undertake a comprehensive review of colorectal and gastric cancer detection specifically employing traditional ML classifiers. This review emphasizes the mathematical underpinnings of cancer detection, encompassing preprocessing techniques, feature extraction, machine learning classifiers, and performance assessment metrics. We provide mathematical formulations for these key components. Our analysis is limited to peer-reviewed articles published between 2017 and 2023, exclusively considering medical imaging datasets. Benchmark and publicly available imaging datasets for colorectal and gastric cancers are presented. This review synthesizes findings from 20 articles on colorectal cancer and 16 on gastric cancer, culminating in a total of 36 research articles. A significant focus is placed on mathematical formulations for commonly used preprocessing techniques, features, ML classifiers, and assessment metrics. Crucially, we introduce our optimized methodology for the detection of both colorectal and gastric cancers. Our performance metrics analysis reveals remarkable results: 100% accuracy in both cancer types, but with the lowest sensitivity recorded at 43.1% for gastric cancer.
Collapse
Affiliation(s)
- Hari Mohan Rai
- School of Computing, Gachon University, Seongnam-si 13120, Republic of Korea
| | - Joon Yoo
- School of Computing, Gachon University, Seongnam-si 13120, Republic of Korea
| |
Collapse
|
19
|
Weerarathna IN, Kamble AR, Luharia A. Artificial Intelligence Applications for Biomedical Cancer Research: A Review. Cureus 2023; 15:e48307. [PMID: 38058345 PMCID: PMC10697339 DOI: 10.7759/cureus.48307] [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: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 12/08/2023] Open
Abstract
Artificial intelligence (AI) has rapidly evolved and demonstrated its potential in transforming biomedical cancer research, offering innovative solutions for cancer diagnosis, treatment, and overall patient care. Over the past two decades, AI has played a pivotal role in revolutionizing various facets of cancer clinical research. In this comprehensive review, we delve into the diverse applications of AI across the cancer care continuum, encompassing radiodiagnosis, radiotherapy, chemotherapy, immunotherapy, targeted therapy, surgery, and nanotechnology. AI has revolutionized cancer diagnosis, enabling early detection and precise characterization through advanced image analysis techniques. In radiodiagnosis, AI-driven algorithms enhance the accuracy of medical imaging, making it an invaluable tool for clinicians in the detection and assessment of cancer. AI has also revolutionized radiotherapy, facilitating precise tumor boundary delineation, optimizing treatment planning, and enabling real-time adjustments to improve therapeutic outcomes while minimizing collateral damage to healthy tissues. In chemotherapy, AI models have emerged as powerful tools for predicting patient responses to different treatment regimens, allowing for more personalized and effective strategies. In immunotherapy, AI analyzes genetic and imaging data to select ideal candidates for treatment and predict responses. Targeted therapy has seen great advancements with AI, aiding in the identification of specific molecular targets for tailored treatments. AI plays a vital role in surgery by offering real-time navigation and support, enhancing surgical precision. Moreover, the synergy between AI and nanotechnology promises the development of personalized nanomedicines, offering more efficient and targeted cancer treatments. While challenges related to data quality, interpretability, and ethical considerations persist, the future of AI in cancer research holds tremendous promise for improving patient outcomes through advanced and individualized care.
Collapse
Affiliation(s)
- Induni N Weerarathna
- Biomedical Sciences, School of Allied Health Sciences, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Aahash R Kamble
- Artificial Intelligence and Data Science, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Anurag Luharia
- Radiotherapy, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
20
|
Rai HM, Yoo J. A comprehensive analysis of recent advancements in cancer detection using machine learning and deep learning models for improved diagnostics. J Cancer Res Clin Oncol 2023; 149:14365-14408. [PMID: 37540254 DOI: 10.1007/s00432-023-05216-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023]
Abstract
PURPOSE There are millions of people who lose their life due to several types of fatal diseases. Cancer is one of the most fatal diseases which may be due to obesity, alcohol consumption, infections, ultraviolet radiation, smoking, and unhealthy lifestyles. Cancer is abnormal and uncontrolled tissue growth inside the body which may be spread to other body parts other than where it has originated. Hence it is very much required to diagnose the cancer at an early stage to provide correct and timely treatment. Also, manual diagnosis and diagnostic error may cause of the death of many patients hence much research are going on for the automatic and accurate detection of cancer at early stage. METHODS In this paper, we have done the comparative analysis of the diagnosis and recent advancement for the detection of various cancer types using traditional machine learning (ML) and deep learning (DL) models. In this study, we have included four types of cancers, brain, lung, skin, and breast and their detection using ML and DL techniques. In extensive review we have included a total of 130 pieces of literature among which 56 are of ML-based and 74 are from DL-based cancer detection techniques. Only the peer reviewed research papers published in the recent 5-year span (2018-2023) have been included for the analysis based on the parameters, year of publication, feature utilized, best model, dataset/images utilized, and best accuracy. We have reviewed ML and DL-based techniques for cancer detection separately and included accuracy as the performance evaluation metrics to maintain the homogeneity while verifying the classifier efficiency. RESULTS Among all the reviewed literatures, DL techniques achieved the highest accuracy of 100%, while ML techniques achieved 99.89%. The lowest accuracy achieved using DL and ML approaches were 70% and 75.48%, respectively. The difference in accuracy between the highest and lowest performing models is about 28.8% for skin cancer detection. In addition, the key findings, and challenges for each type of cancer detection using ML and DL techniques have been presented. The comparative analysis between the best performing and worst performing models, along with overall key findings and challenges, has been provided for future research purposes. Although the analysis is based on accuracy as the performance metric and various parameters, the results demonstrate a significant scope for improvement in classification efficiency. CONCLUSION The paper concludes that both ML and DL techniques hold promise in the early detection of various cancer types. However, the study identifies specific challenges that need to be addressed for the widespread implementation of these techniques in clinical settings. The presented results offer valuable guidance for future research in cancer detection, emphasizing the need for continued advancements in ML and DL-based approaches to improve diagnostic accuracy and ultimately save more lives.
Collapse
Affiliation(s)
- Hari Mohan Rai
- School of Computing, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, 13120, Gyeonggi-do, Republic of Korea.
| | - Joon Yoo
- School of Computing, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
21
|
Martins JC, Maier J, Gianoli C, Neppl S, Dedes G, Alhazmi A, Veloza S, Reiner M, Belka C, Kachelrieß M, Parodi K. Towards real-time EPID-based 3D in vivo dosimetry for IMRT with Deep Neural Networks: A feasibility study. Phys Med 2023; 114:103148. [PMID: 37801811 DOI: 10.1016/j.ejmp.2023.103148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 08/17/2023] [Accepted: 09/22/2023] [Indexed: 10/08/2023] Open
Abstract
We investigate the potential of the Deep Dose Estimate (DDE) neural network to predict 3D dose distributions inside patients with Monte Carlo (MC) accuracy, based on transmitted EPID signals and patient CTs. The network was trained using as input patient CTs and first-order dose approximations (FOD). Accurate dose distributions (ADD) simulated with MC were given as training targets. 83 pelvic CTs were used to simulate ADDs and respective EPID signals for subfields of prostate IMRT plans (gantry at 0∘). FODs were produced as backprojections from the EPID signals. 581 ADD-FOD sets were produced and divided into training and test sets. An additional dataset simulated with gantry at 90∘ (lateral set) was used for evaluating the performance of the DDE at different beam directions. The quality of the FODs and DDE-predicted dose distributions (DDEP) with respect to ADDs, from the test and lateral sets, was evaluated with gamma analysis (3%,2 mm). The passing rates between FODs and ADDs were as low as 46%, while for DDEPs the passing rates were above 97% for the test set. Meaningful improvements were also observed for the lateral set. The high passing rates for DDEPs indicate that the DDE is able to convert FODs into ADDs. Moreover, the trained DDE predicts the dose inside a patient CT within 0.6 s/subfield (GPU), in contrast to 14 h needed for MC (CPU-cluster). 3D in vivo dose distributions due to clinical patient irradiation can be obtained within seconds, with MC-like accuracy, potentially paving the way towards real-time EPID-based in vivo dosimetry.
Collapse
Affiliation(s)
- Juliana Cristina Martins
- Department of Medical Physics, Faculty of Physics, Ludwig-Maximilians-Universität München, Am Coulombwall 1, Garching b. München, 85748, Germany.
| | - Joscha Maier
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, 69120, Germany.
| | - Chiara Gianoli
- Department of Medical Physics, Faculty of Physics, Ludwig-Maximilians-Universität München, Am Coulombwall 1, Garching b. München, 85748, Germany.
| | - Sebastian Neppl
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, Munich, 81377, Germany.
| | - George Dedes
- Department of Medical Physics, Faculty of Physics, Ludwig-Maximilians-Universität München, Am Coulombwall 1, Garching b. München, 85748, Germany.
| | - Abdulaziz Alhazmi
- Department of Medical Physics, Faculty of Physics, Ludwig-Maximilians-Universität München, Am Coulombwall 1, Garching b. München, 85748, Germany.
| | - Stella Veloza
- Department of Medical Physics, Faculty of Physics, Ludwig-Maximilians-Universität München, Am Coulombwall 1, Garching b. München, 85748, Germany.
| | - Michael Reiner
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, Munich, 81377, Germany.
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistraße 15, Munich, 81377, Germany.
| | - Marc Kachelrieß
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, 69120, Germany; Heidelberg University, Grabengasse 1, Heidelberg, 69117, Germany.
| | - Katia Parodi
- Department of Medical Physics, Faculty of Physics, Ludwig-Maximilians-Universität München, Am Coulombwall 1, Garching b. München, 85748, Germany.
| |
Collapse
|
22
|
Kuo CY, Kuo LJ, Lin YK. Artificial intelligence based system for predicting permanent stoma after sphincter saving operations. Sci Rep 2023; 13:16039. [PMID: 37749194 PMCID: PMC10519982 DOI: 10.1038/s41598-023-43211-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023] Open
Abstract
Although the goal of rectal cancer treatment is to restore gastrointestinal continuity, some patients with rectal cancer develop a permanent stoma (PS) after sphincter-saving operations. Although many studies have identified the risk factors and causes of PS, few have precisely predicted the probability of PS formation before surgery. To validate whether an artificial intelligence model can accurately predict PS formation in patients with rectal cancer after sphincter-saving operations. Patients with rectal cancer who underwent a sphincter-saving operation at Taipei Medical University Hospital between January 1, 2012, and December 31, 2021, were retrospectively included in this study. A machine learning technique was used to predict whether a PS would form after a sphincter-saving operation. We included 19 routinely available preoperative variables in the artificial intelligence analysis. To evaluate the efficiency of the model, 6 performance metrics were utilized: accuracy, sensitivity, specificity, positive predictive value, negative predictive value, and area under the receiving operating characteristic curve. In our classification pipeline, the data were randomly divided into a training set (80% of the data) and a validation set (20% of the data). The artificial intelligence models were trained using the training dataset, and their performance was evaluated using the validation dataset. Synthetic minority oversampling was used to solve the data imbalance. A total of 428 patients were included, and the PS rate was 13.6% (58/428) in the training set. The logistic regression (LR), Gaussian Naïve Bayes (GNB), Extreme Gradient Boosting (XGB), Gradient Boosting (GB), random forest, decision tree and light gradient boosting machine (LightGBM) algorithms were employed. The accuracies of the logistic regression (LR), Gaussian Naïve Bayes (GNB), Extreme Gradient Boosting (XGB), Gradient Boosting (GB), random forest (RF), decision tree (DT) and light gradient boosting machine (LightGBM) models were 70%, 76%, 89%, 93%, 95%, 79% and 93%, respectively. The area under the receiving operating characteristic curve values were 0.79 for the LR model, 0.84 for the GNB, 0.95 for the XGB, 0.95 for the GB, 0.99 for the RF model, 0.79 for the DT model and 0.98 for the LightGBM model. The key predictors that were identified were the distance of the lesion from the anal verge, clinical N stage, age, sex, American Society of Anesthesiologists score, and preoperative albumin and carcinoembryonic antigen levels. Integration of artificial intelligence with available preoperative data can potentially predict stoma outcomes after sphincter-saving operations. Our model exhibited excellent predictive ability and can improve the process of obtaining informed consent.
Collapse
Affiliation(s)
- Chih-Yu Kuo
- Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Li-Jen Kuo
- Division of Colorectal Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
| | - Yen-Kuang Lin
- Graduate Institute of Athletics and Coaching Science, National Taiwan Sport University, Taoyuan, Taiwan.
| |
Collapse
|
23
|
Kamalanathan A, Muthu B, Kuniyil Kaleena P. Artificial Intelligence (AI) Game Changer in Cancer Biology. MARVELS OF ARTIFICIAL AND COMPUTATIONAL INTELLIGENCE IN LIFE SCIENCES 2023:62-87. [DOI: 10.2174/9789815136807123010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Healthcare is one of many industries where the most modern technologies,
such as artificial intelligence and machine learning, have shown a wide range of
applications. Cancer, one of the most prevalent non-communicable diseases in modern
times, accounts for a sizable portion of worldwide mortality. Investigations are
continuously being conducted to find ways to reduce cancer mortality and morbidity.
Artificial Intelligence (AI) is currently being used in cancer research, with promising
results. Two main features play a vital role in improving cancer prognosis: early
detection and proper diagnosis using imaging and molecular techniques. AI's use as a
tool in these sectors has demonstrated its capacity to precisely detect and diagnose,
which is one of AI's many applications in cancer research. The purpose of this chapter
is to review the literature and find AI applications in a range of cancers that are
commonly seen.
Collapse
Affiliation(s)
- Ashok Kamalanathan
- Department of Microbiology and Biotechnology, Faculty of Arts and Science, Bharath Institute
of Higher Education and Research (BIHER), Chennai- 600 073, Tamil Nadu, India
| | - Babu Muthu
- Department of Microbiology and Biotechnology, Faculty of Arts and Science, Bharath Institute
of Higher Education and Research (BIHER), Chennai- 600 073, Tamil Nadu, India
| | | |
Collapse
|
24
|
Swillens JEM, Nagtegaal ID, Engels S, Lugli A, Hermens RPMG, van der Laak JAWM. Pathologists' first opinions on barriers and facilitators of computational pathology adoption in oncological pathology: an international study. Oncogene 2023; 42:2816-2827. [PMID: 37587332 PMCID: PMC10504072 DOI: 10.1038/s41388-023-02797-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 07/26/2023] [Indexed: 08/18/2023]
Abstract
Computational pathology (CPath) algorithms detect, segment or classify cancer in whole slide images, approaching or even exceeding the accuracy of pathologists. Challenges have to be overcome before these algorithms can be used in practice. We therefore aim to explore international perspectives on the future role of CPath in oncological pathology by focusing on opinions and first experiences regarding barriers and facilitators. We conducted an international explorative eSurvey and semi-structured interviews with pathologists utilizing an implementation framework to classify potential influencing factors. The eSurvey results showed remarkable variation in opinions regarding attitude, understandability and validation of CPath. Interview results showed that barriers focused on the quality of available evidence, while most facilitators concerned strengths of CPath. A lack of consensus was present for multiple factors, such as the determination of sufficient validation using CPath, the preferred function of CPath within the digital workflow and the timing of CPath introduction in pathology education. The diversity in opinions illustrates variety in influencing factors in CPath adoption. A next step would be to quantitatively determine important factors for adoption and initiate validation studies. Both should include clear case descriptions and be conducted among a more homogenous panel of pathologists based on sub specialization.
Collapse
Affiliation(s)
- Julie E M Swillens
- Scientific Center for Quality of Healthcare (IQ Healthcare), Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - Iris D Nagtegaal
- Department of Pathology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Sam Engels
- Scientific Center for Quality of Healthcare (IQ Healthcare), Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | - Rosella P M G Hermens
- Scientific Center for Quality of Healthcare (IQ Healthcare), Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | | |
Collapse
|
25
|
Williams AH, Zhan CG. Staying Ahead of the Game: How SARS-CoV-2 has Accelerated the Application of Machine Learning in Pandemic Management. BioDrugs 2023; 37:649-674. [PMID: 37464099 DOI: 10.1007/s40259-023-00611-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2023] [Indexed: 07/20/2023]
Abstract
In recent years, machine learning (ML) techniques have garnered considerable interest for their potential use in accelerating the rate of drug discovery. With the emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, the utilization of ML has become even more crucial in the search for effective antiviral medications. The pandemic has presented the scientific community with a unique challenge, and the rapid identification of potential treatments has become an urgent priority. Researchers have been able to accelerate the process of identifying drug candidates, repurposing existing drugs, and designing new compounds with desirable properties using machine learning in drug discovery. To train predictive models, ML techniques in drug discovery rely on the analysis of large datasets, including both experimental and clinical data. These models can be used to predict the biological activities, potential side effects, and interactions with specific target proteins of drug candidates. This strategy has proven to be an effective method for identifying potential coronavirus disease 2019 (COVID-19) and other disease treatments. This paper offers a thorough analysis of the various ML techniques implemented to combat COVID-19, including supervised and unsupervised learning, deep learning, and natural language processing. The paper discusses the impact of these techniques on pandemic drug development, including the identification of potential treatments, the understanding of the disease mechanism, and the creation of effective and safe therapeutics. The lessons learned can be applied to future outbreaks and drug discovery initiatives.
Collapse
Affiliation(s)
- Alexander H Williams
- Molecular Modeling and Biopharmaceutical Center, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
- GSK Upper Providence, 1250 S. Collegeville Road, Collegeville, PA, 19426, USA
| | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA.
| |
Collapse
|
26
|
Swanson K, Wu E, Zhang A, Alizadeh AA, Zou J. From patterns to patients: Advances in clinical machine learning for cancer diagnosis, prognosis, and treatment. Cell 2023; 186:1772-1791. [PMID: 36905928 DOI: 10.1016/j.cell.2023.01.035] [Citation(s) in RCA: 185] [Impact Index Per Article: 92.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/10/2023] [Accepted: 01/26/2023] [Indexed: 03/12/2023]
Abstract
Machine learning (ML) is increasingly used in clinical oncology to diagnose cancers, predict patient outcomes, and inform treatment planning. Here, we review recent applications of ML across the clinical oncology workflow. We review how these techniques are applied to medical imaging and to molecular data obtained from liquid and solid tumor biopsies for cancer diagnosis, prognosis, and treatment design. We discuss key considerations in developing ML for the distinct challenges posed by imaging and molecular data. Finally, we examine ML models approved for cancer-related patient usage by regulatory agencies and discuss approaches to improve the clinical usefulness of ML.
Collapse
Affiliation(s)
- Kyle Swanson
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Eric Wu
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Angela Zhang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ash A Alizadeh
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - James Zou
- Department of Computer Science, Stanford University, Stanford, CA, USA; Department of Electrical Engineering, Stanford University, Stanford, CA, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
| |
Collapse
|
27
|
Xuan A, Chen H, Chen T, Li J, Lu S, Fan T, Zeng D, Wen Z, Ma J, Hunter D, Ding C, Zhu Z. The application of machine learning in early diagnosis of osteoarthritis: a narrative review. Ther Adv Musculoskelet Dis 2023; 15:1759720X231158198. [PMID: 36937823 PMCID: PMC10017946 DOI: 10.1177/1759720x231158198] [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: 09/02/2022] [Accepted: 02/01/2023] [Indexed: 03/16/2023] Open
Abstract
Osteoarthritis (OA) is the commonest musculoskeletal disease worldwide, with an increasing prevalence due to aging. It causes joint pain and disability, decreased quality of life, and a huge burden on healthcare services for society. However, the current main diagnostic methods are not suitable for early diagnosing patients of OA. The use of machine learning (ML) in OA diagnosis has increased dramatically in the past few years. Hence, in this review article, we describe the research progress in the application of ML in the early diagnosis of OA, discuss the current trends and limitations of ML approaches, and propose future research priorities to apply the tools in the field of OA. Accurate ML-based predictive models with imaging techniques that are sensitive to early changes in OA ahead of the emergence of clinical features are expected to address the current dilemma. The diagnostic ability of the fusion model that combines multidimensional information makes patient-specific early diagnosis and prognosis estimation of OA possible in the future.
Collapse
Affiliation(s)
- Anran Xuan
- The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haowei Chen
- The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tianyu Chen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jia Li
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nafang Hospital, Southern Medical University, Guangzhou, China
| | - Shilong Lu
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tianxiang Fan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dong Zeng
- College of Automation Science and Engineering, South China University of Technology, Guangzhou, China
| | - Zhibo Wen
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianhua Ma
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - David Hunter
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology, Royal North Shore Hospital and Institute of Bone and Joint Research, Kolling Institute, University of Sydney, Sydney, NSW, Australia
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, 261 Industry Road, Guangzhou, 510280, China
- Department of Rheumatology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Orthopaedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
28
|
Yang Y, Zhang Y, Li Y. Artificial intelligence applications in pediatric oncology diagnosis. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:157-169. [PMID: 36937318 PMCID: PMC10017189 DOI: 10.37349/etat.2023.00127] [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: 10/17/2022] [Accepted: 12/30/2022] [Indexed: 03/04/2023] Open
Abstract
Artificial intelligence (AI) algorithms have been applied in abundant medical tasks with high accuracy and efficiency. Physicians can improve their diagnostic efficiency with the assistance of AI techniques for improving the subsequent personalized treatment and surveillance. AI algorithms fundamentally capture data, identify underlying patterns, achieve preset endpoints, and provide decisions and predictions about real-world events with working principles of machine learning and deep learning. AI algorithms with sufficient graphic processing unit power have been demonstrated to provide timely diagnostic references based on preliminary training of large amounts of clinical and imaging data. The sample size issue is an inevitable challenge for pediatric oncology considering its low morbidity and individual heterogeneity. However, this problem may be solved in the near future considering the exponential advancements of AI algorithms technically to decrease the dependence of AI operation on the amount of data sets and the efficiency of computing power. For instance, it could be a feasible solution by shifting convolutional neural networks (CNNs) from adults and sharing CNN algorithms across multiple institutions besides original data. The present review provides important insights into emerging AI applications for the diagnosis of pediatric oncology by systematically overviewing of up-to-date literature.
Collapse
Affiliation(s)
- Yuhan Yang
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yimao Zhang
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuan Li
- Laboratory of Digestive Surgery, State Key Laboratory of Biotherapy and Cancer Center, Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
29
|
Predicting gene mutation status via artificial intelligence technologies based on multimodal integration (MMI) to advance precision oncology. Semin Cancer Biol 2023; 91:1-15. [PMID: 36801447 DOI: 10.1016/j.semcancer.2023.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/30/2023] [Accepted: 02/15/2023] [Indexed: 02/21/2023]
Abstract
Personalized treatment strategies for cancer frequently rely on the detection of genetic alterations which are determined by molecular biology assays. Historically, these processes typically required single-gene sequencing, next-generation sequencing, or visual inspection of histopathology slides by experienced pathologists in a clinical context. In the past decade, advances in artificial intelligence (AI) technologies have demonstrated remarkable potential in assisting physicians with accurate diagnosis of oncology image-recognition tasks. Meanwhile, AI techniques make it possible to integrate multimodal data such as radiology, histology, and genomics, providing critical guidance for the stratification of patients in the context of precision therapy. Given that the mutation detection is unaffordable and time-consuming for a considerable number of patients, predicting gene mutations based on routine clinical radiological scans or whole-slide images of tissue with AI-based methods has become a hot issue in actual clinical practice. In this review, we synthesized the general framework of multimodal integration (MMI) for molecular intelligent diagnostics beyond standard techniques. Then we summarized the emerging applications of AI in the prediction of mutational and molecular profiles of common cancers (lung, brain, breast, and other tumor types) pertaining to radiology and histology imaging. Furthermore, we concluded that there truly exist multiple challenges of AI techniques in the way of its real-world application in the medical field, including data curation, feature fusion, model interpretability, and practice regulations. Despite these challenges, we still prospect the clinical implementation of AI as a highly potential decision-support tool to aid oncologists in future cancer treatment management.
Collapse
|
30
|
Pan J, Hong G, Zeng H, Liao C, Li H, Yao Y, Gan Q, Wang Y, Wu S, Lin T. An artificial intelligence model for the pathological diagnosis of invasion depth and histologic grade in bladder cancer. J Transl Med 2023; 21:42. [PMID: 36691055 PMCID: PMC9869632 DOI: 10.1186/s12967-023-03888-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 01/12/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Accurate pathological diagnosis of invasion depth and histologic grade is key for clinical management in patients with bladder cancer (BCa), but it is labour-intensive, experience-dependent and subject to interobserver variability. Here, we aimed to develop a pathological artificial intelligence diagnostic model (PAIDM) for BCa diagnosis. METHODS A total of 854 whole slide images (WSIs) from 692 patients were included and divided into training and validation sets. The PAIDM was developed using the training set based on the deep learning algorithm ScanNet, and the performance was verified at the patch level in validation set 1 and at the WSI level in validation set 2. An independent validation cohort (validation set 3) was employed to compare the PAIDM and pathologists. Model performance was evaluated using the area under the curve (AUC), accuracy, sensitivity, specificity, positive predictive value and negative predictive value. RESULTS The AUCs of the PAIDM were 0.878 (95% CI 0.875-0.881) at the patch level in validation set 1 and 0.870 (95% CI 0.805-0.923) at the WSI level in validation set 2. In comparing the PAIDM and pathologists, the PAIDM achieved an AUC of 0.847 (95% CI 0.779-0.905), which was non-inferior to the average diagnostic level of pathologists. There was high consistency between the model-predicted and manually annotated areas, improving the PAIDM's interpretability. CONCLUSIONS We reported an artificial intelligence-based diagnostic model for BCa that performed well in identifying invasion depth and histologic grade. Importantly, the PAIDM performed admirably in patch-level recognition, with a promising application for transurethral resection specimens.
Collapse
Affiliation(s)
- Jiexin Pan
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guibin Hong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong Zeng
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chengxiao Liao
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
| | - Huarun Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
| | - Yuhui Yao
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
| | - Qinghua Gan
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
| | - Yun Wang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
| | - Shaoxu Wu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, China.
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, China.
| |
Collapse
|
31
|
Juhas M. Artificial Intelligence in Microbiology. BRIEF LESSONS IN MICROBIOLOGY 2023:93-109. [DOI: 10.1007/978-3-031-29544-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
32
|
Deep learning for preoperative prediction of the EGFR mutation and subtypes based on the MRI image of spinal metastasis from primary NSCLC. Biomed Signal Process Control 2023. [DOI: 10.1016/j.bspc.2022.104084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
33
|
Artificial intelligence for prediction of response to cancer immunotherapy. Semin Cancer Biol 2022; 87:137-147. [PMID: 36372326 DOI: 10.1016/j.semcancer.2022.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Artificial intelligence (AI) indicates the application of machines to imitate intelligent behaviors for solving complex tasks with minimal human intervention, including machine learning and deep learning. The use of AI in medicine improves health-care systems in multiple areas such as diagnostic confirmation, risk stratification, analysis, prognosis prediction, treatment surveillance, and virtual health support, which has considerable potential to revolutionize and reshape medicine. In terms of immunotherapy, AI has been applied to unlock underlying immune signatures to associate with responses to immunotherapy indirectly as well as predict responses to immunotherapy responses directly. The AI-based analysis of high-throughput sequences and medical images can provide useful information for management of cancer immunotherapy considering the excellent abilities in selecting appropriate subjects, improving therapeutic regimens, and predicting individualized prognosis. In present review, we aim to evaluate a broad framework about AI-based computational approaches for prediction of response to cancer immunotherapy on both indirect and direct manners. Furthermore, we summarize our perspectives about challenges and opportunities of further AI applications on cancer immunotherapy relating to clinical practicability.
Collapse
|
34
|
Leary D, Basran PS. The role of artificial intelligence in veterinary radiation oncology. Vet Radiol Ultrasound 2022; 63 Suppl 1:903-912. [PMID: 36514233 DOI: 10.1111/vru.13162] [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: 08/20/2021] [Revised: 01/21/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022] Open
Abstract
Veterinary radiation oncology regularly deploys sophisticated contouring, image registration, and treatment planning optimization software for patient care. Over the past decade, advances in computing power and the rapid development of neural networks, open-source software packages, and data science have been realized and resulted in new research and clinical applications of artificial intelligent (AI) systems in radiation oncology. These technologies differ from conventional software in their level of complexity and ability to learn from representative and local data. We provide clinical and research application examples of AI in human radiation oncology and their potential applications in veterinary medicine throughout the patient's care-path: from treatment simulation, deformable registration, auto-segmentation, automated treatment planning and plan selection, quality assurance, adaptive radiotherapy, and outcomes modeling. These technologies have the potential to offer significant time and cost savings in the veterinary setting; however, since the range of usefulness of these technologies have not been well studied nor understood, care must be taken if adopting AI technologies in clinical practice. Over the next several years, some practical and realizable applications of AI in veterinary radiation oncology include automated segmentation of normal tissues and tumor volumes, deformable registration, multi-criteria plan optimization, and adaptive radiotherapy. Keys in achieving success in adopting AI in veterinary radiation oncology include: establishing "truth-data"; data harmonization; multi-institutional data and collaborations; standardized dose reporting and taxonomy; adopting an open access philosophy, data collection and curation; open-source algorithm development; and transparent and platform-independent code development.
Collapse
Affiliation(s)
- Del Leary
- Department of Environment and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Parminder S Basran
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
35
|
Adeoye J, Akinshipo A, Koohi-Moghadam M, Thomson P, Su YX. Construction of machine learning-based models for cancer outcomes in low and lower-middle income countries: A scoping review. Front Oncol 2022; 12:976168. [PMID: 36531037 PMCID: PMC9751812 DOI: 10.3389/fonc.2022.976168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/14/2022] [Indexed: 01/31/2025] Open
Abstract
Background The impact and utility of machine learning (ML)-based prediction tools for cancer outcomes including assistive diagnosis, risk stratification, and adjunctive decision-making have been largely described and realized in the high income and upper-middle-income countries. However, statistical projections have estimated higher cancer incidence and mortality risks in low and lower-middle-income countries (LLMICs). Therefore, this review aimed to evaluate the utilization, model construction methods, and degree of implementation of ML-based models for cancer outcomes in LLMICs. Methods PubMed/Medline, Scopus, and Web of Science databases were searched and articles describing the use of ML-based models for cancer among local populations in LLMICs between 2002 and 2022 were included. A total of 140 articles from 22,516 citations that met the eligibility criteria were included in this study. Results ML-based models from LLMICs were often based on traditional ML algorithms than deep or deep hybrid learning. We found that the construction of ML-based models was skewed to particular LLMICs such as India, Iran, Pakistan, and Egypt with a paucity of applications in sub-Saharan Africa. Moreover, models for breast, head and neck, and brain cancer outcomes were frequently explored. Many models were deemed suboptimal according to the Prediction model Risk of Bias Assessment tool (PROBAST) due to sample size constraints and technical flaws in ML modeling even though their performance accuracy ranged from 0.65 to 1.00. While the development and internal validation were described for all models included (n=137), only 4.4% (6/137) have been validated in independent cohorts and 0.7% (1/137) have been assessed for clinical impact and efficacy. Conclusion Overall, the application of ML for modeling cancer outcomes in LLMICs is increasing. However, model development is largely unsatisfactory. We recommend model retraining using larger sample sizes, intensified external validation practices, and increased impact assessment studies using randomized controlled trial designs. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=308345, identifier CRD42022308345.
Collapse
Affiliation(s)
- John Adeoye
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Hong Kong, Hong Kong, Hong Kong, SAR, China
- Oral Cancer Research Theme, Faculty of Dentistry, University of Hong Kong, Hong Kong, Hong Kong, SAR, China
| | - Abdulwarith Akinshipo
- Department of Oral and Maxillofacial Pathology and Biology, Faculty of Dentistry, University of Lagos, Lagos, Nigeria
| | - Mohamad Koohi-Moghadam
- Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, University of Hong Kong, Hong Kong, Hong Kong, SAR, China
- Clinical Artificial Intelligence Research Theme, Faculty of Dentistry, University of Hong Kong, Hong Kong, Hong Kong, SAR, China
| | - Peter Thomson
- College of Medicine and Dentistry, James Cook University, Cairns, Queensland, Australia
| | - Yu-Xiong Su
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Hong Kong, Hong Kong, Hong Kong, SAR, China
- Oral Cancer Research Theme, Faculty of Dentistry, University of Hong Kong, Hong Kong, Hong Kong, SAR, China
| |
Collapse
|
36
|
Cordier BA, Sawaya NPD, Guerreschi GG, McWeeney SK. Biology and medicine in the landscape of quantum advantages. J R Soc Interface 2022; 19:20220541. [PMID: 36448288 PMCID: PMC9709576 DOI: 10.1098/rsif.2022.0541] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/04/2022] [Indexed: 12/03/2022] Open
Abstract
Quantum computing holds substantial potential for applications in biology and medicine, spanning from the simulation of biomolecules to machine learning methods for subtyping cancers on the basis of clinical features. This potential is encapsulated by the concept of a quantum advantage, which is contingent on a reduction in the consumption of a computational resource, such as time, space or data. Here, we distill the concept of a quantum advantage into a simple framework to aid researchers in biology and medicine pursuing the development of quantum applications. We then apply this framework to a wide variety of computational problems relevant to these domains in an effort to (i) assess the potential of practical advantages in specific application areas and (ii) identify gaps that may be addressed with novel quantum approaches. In doing so, we provide an extensive survey of the intersection of biology and medicine with the current landscape of quantum algorithms and their potential advantages. While we endeavour to identify specific computational problems that may admit practical advantages throughout this work, the rapid pace of change in the fields of quantum computing, classical algorithms and biological research implies that this intersection will remain highly dynamic for the foreseeable future.
Collapse
Affiliation(s)
- Benjamin A. Cordier
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, OR 97202, USA
| | | | | | - Shannon K. McWeeney
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, OR 97202, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97202, USA
- Oregon Clinical and Translational Research Institute, Oregon Health and Science University, Portland, OR 97202, USA
| |
Collapse
|
37
|
Muacevic A, Adler JR. Role of Artificial Intelligence and Machine Learning in Prediction, Diagnosis, and Prognosis of Cancer. Cureus 2022; 14:e31008. [PMID: 36475188 PMCID: PMC9717523 DOI: 10.7759/cureus.31008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/02/2022] [Indexed: 01/25/2023] Open
Abstract
Cancer is one of the most devastating, fatal, dangerous, and unpredictable ailments. To reduce the risk of fatality in this disease, we need some ways to predict the disease, diagnose it faster and precisely, and predict the prognosis accurately. The incorporation of artificial intelligence (AI), machine learning (ML), and deep learning (DL) algorithms into the healthcare system has already proven to work wonders for patients. Artificial intelligence is a simulation of intelligence that uses data, rules, and information programmed in it to make predictions. The science of machine learning (ML) uses data to enhance performance in a variety of activities and tasks. A bigger family of machine learning techniques built on artificial neural networks and representation learning is deep learning (DL). To clarify, we require AI, ML, and DL to predict cancer risk, survival chances, cancer recurrence, cancer diagnosis, and cancer prognosis. All of these are required to improve patient's quality of life, increase their survival rates, decrease anxiety and fear to some extent, and make a proper personalized treatment plan for the suffering patient. The survival rates of people with diffuse large B-cell lymphoma (DLBCL) can be forecasted. Both solid and non-solid tumors can be diagnosed precisely with the help of AI and ML algorithms. The prognosis of the disease can also be forecasted with AI and its approaches like deep learning. This improvement in cancer care is a turning point in advanced healthcare and will deeply impact patient's life for good.
Collapse
|
38
|
Development and validation of a combined nomogram model based on deep learning contrast-enhanced ultrasound and clinical factors to predict preoperative aggressiveness in pancreatic neuroendocrine neoplasms. Eur Radiol 2022; 32:7965-7975. [PMID: 35389050 DOI: 10.1007/s00330-022-08703-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/21/2022] [Accepted: 02/25/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVES This study aimed to develop and validate a combined nomogram model based on deep learning (DL) contrast-enhanced ultrasound (CEUS) and clinical factors to preoperatively predict the aggressiveness of pancreatic neuroendocrine neoplasms (PNENs). METHODS In this retrospective study, consecutive patients with histologically proven PNENs underwent CEUS examination at the initial work-up between January 2010 and October 2020. Patients were randomly allocated to the training and test sets. Typical sonographic and enhanced images of PNENs were selected to fine-tune the SE-ResNeXt-50 network. A combined nomogram model was developed by incorporating the DL predictive probability with clinical factors using multivariate logistic regression analysis. The utility of the proposed model was evaluated using receiver operator characteristic, calibration, and decision curve analysis. RESULTS A total of 104 patients were evaluated, including 80 (mean age ± standard deviation, 47 years ± 12; 56 males) in the training set and 24 (50 years ± 12; 14 males) in the test set. The DL model displayed effective image recognition with an AUC of 0.81 (95%CI: 0.62-1.00) in the test set. The combined nomogram model that incorporated independent clinical risk factors, such as tumor size, arterial enhancement level, and DL predictive probability, showed strong discrimination, with an AUC of 0.85 (95%CI: 0.69-1.00) in the test set with good calibration. Decision curve analysis verified the clinical usefulness of the combined nomogram. CONCLUSIONS The combined nomogram model could serve as a preoperative, noninvasive, and precise evaluation tool to differentiate aggressive and non-aggressive PNENs. KEY POINTS • Tumor size (odds ratio [OR], 1.58; p = 0.02), arterial enhancement level (OR, 0.04; p = 0.008), and deep learning predictive probability (OR, 288.46; p < 0.001) independently predicted aggressiveness of pancreatic neuroendocrine neoplasms preoperatively. • The combined model predicted aggressiveness better than the clinical model (AUC: 0.97 vs. 0.87, p = 0.009), achieving AUC values of 0.97 and 0.85 in the training set and the test set, respectively.
Collapse
|
39
|
Manganelli Conforti P, D’Acunto M, Russo P. Deep Learning for Chondrogenic Tumor Classification through Wavelet Transform of Raman Spectra. SENSORS (BASEL, SWITZERLAND) 2022; 22:s22197492. [PMID: 36236597 PMCID: PMC9571786 DOI: 10.3390/s22197492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 05/22/2023]
Abstract
The grading of cancer tissues is still one of the main challenges for pathologists. The development of enhanced analysis strategies hence becomes crucial to accurately identify and further deal with each individual case. Raman spectroscopy (RS) is a promising tool for the classification of tumor tissues as it allows us to obtain the biochemical maps of the tissues under analysis and to observe their evolution in terms of biomolecules, proteins, lipid structures, DNA, vitamins, and so on. However, its potential could be further improved by providing a classification system which would be able to recognize the sample tumor category by taking as input the raw Raman spectroscopy signal; this could provide more reliable responses in shorter time scales and could reduce or eliminate false-positive or -negative diagnoses. Deep Learning techniques have become ubiquitous in recent years, with models able to perform classification with high accuracy in most diverse fields of research, e.g., natural language processing, computer vision, medical imaging. However, deep models often rely on huge labeled datasets to produce reasonable accuracy, otherwise occurring in overfitting issues when the training data is insufficient. In this paper, we propose a chondrogenic tumor CLAssification through wavelet transform of RAman spectra (CLARA), which is able to classify with high accuracy Raman spectra obtained from bone tissues. CLARA recognizes and grades the tumors in the evaluated dataset with 97% accuracy by exploiting a classification pipeline consisting of the division of the original task in two binary classification steps, where the first is performed on the original RS signals while the latter is accomplished through the use of a hybrid temporal-frequency 2D transform.
Collapse
Affiliation(s)
| | - Mario D’Acunto
- CNR-IBF, Istituto di Biofisica, Via Moruzzi 1, 56124 Pisa, Italy
| | - Paolo Russo
- DIAG Department, Sapienza University of Rome, Via Ariosto 25, 00185 Roma, Italy
- Correspondence:
| |
Collapse
|
40
|
Tsimberidou AM, Fountzilas E, Bleris L, Kurzrock R. Transcriptomics and solid tumors: The next frontier in precision cancer medicine. Semin Cancer Biol 2022; 84:50-59. [PMID: 32950605 PMCID: PMC11927324 DOI: 10.1016/j.semcancer.2020.09.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 08/16/2020] [Accepted: 09/09/2020] [Indexed: 01/08/2023]
Abstract
Transcriptomics, which encompasses assessments of alternative splicing and alternative polyadenylation, identification of fusion transcripts, explorations of noncoding RNAs, transcript annotation, and discovery of novel transcripts, is a valuable tool for understanding cancer mechanisms and identifying biomarkers. Recent advances in high-throughput technologies have enabled large-scale gene expression profiling. Importantly, RNA expression profiling of tumor tissue has been successfully used to determine clinically actionable molecular alterations. The WINTHER precision medicine clinical trial was the first prospective trial in diverse solid malignancies that assessed both genomics and transcriptomics to match treatments to specific molecular alterations. The use of transcriptome analysis in WINTHER and other trials increased the number of targetable -omic changes compared to genomic profiling alone. Other applications of transcriptomics involve the evaluation of tumor and circulating noncoding RNAs as predictive and prognostic biomarkers, the improvement of risk stratification by the use of prognostic and predictive multigene assays, the identification of fusion transcripts that drive tumors, and an improved understanding of the impact of DNA changes as some genomic alterations are silenced at the RNA level. Finally, RNA sequencing and gene expression analysis have been incorporated into clinical trials to identify markers predicting response to immunotherapy. Many issues regarding the complexity of the analysis, its reproducibility and variability, and the interpretation of the results still need to be addressed. The integration of transcriptomics with genomics, proteomics, epigenetics, and tumor immune profiling will improve biomarker discovery and our understanding of disease mechanisms and, thereby, accelerate the implementation of precision oncology.
Collapse
Affiliation(s)
- Apostolia M Tsimberidou
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX, USA.
| | - Elena Fountzilas
- Department of Medical Oncology, Euromedica General Clinic, Thessaloniki, Greece
| | - Leonidas Bleris
- Bioengineering Department, The University of Texas at Dallas, Richardson, TX, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, CA, USA
| |
Collapse
|
41
|
Application of Deep Learning on the Prognosis of Cutaneous Melanoma Based on Full Scan Pathology Images. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4864485. [PMID: 36072469 PMCID: PMC9441353 DOI: 10.1155/2022/4864485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/13/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022]
Abstract
Introduction The purpose of this study is to use deep learning and machine learning to learn and classify patients with cutaneous melanoma with different prognoses and to explore the application value of deep learning in the prognosis of cutaneous melanoma patients. Methods In deep learning, VGG-19 is selected as the network architecture and learning model for learning and classification. In machine learning, deep features are extracted through the VGG-19 network architecture, and the support vector machine (SVM) model is selected for learning and classification. Compare and explore the application value of deep learning and machine learning in predicting the prognosis of patients with cutaneous melanoma. Result According to receiver operating characteristic (ROC) curves and area under the curve (AUC), the average accuracy of deep learning is higher than that of machine learning, and even the lowest accuracy is better than that of machine learning. Conclusion As the number of learning increases, the accuracy of machine learning and deep learning will increase, but in the same number of cutaneous melanoma patient pathology maps, the accuracy of deep learning will be higher. This study provides new ideas and theories for computational pathology in predicting the prognosis of patients with cutaneous melanoma.
Collapse
|
42
|
Yang Z, Zhu L, Chen Z, Huang M, Ono N, Altaf-Ul-Amin MD, Kanaya S. Cancer Subtyping via Embedded Unsupervised Learning on Transcriptomics Data. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:1113-1116. [PMID: 36085834 DOI: 10.1109/embc48229.2022.9870903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cancer is one of the deadliest diseases worldwide. Accurate diagnosis and classification of cancer subtypes are indispensable for effective clinical treatment. Promising results on automatic cancer subtyping systems have been published recently with the emergence of various deep learning methods. However, such automatic systems often overfit the data due to the high dimensionality and scarcity. In this paper, we propose to investigate automatic subtyping from an unsupervised learning perspective by directly constructing the underlying data distribution itself, hence sufficient data can be generated to alleviate the issue of overfitting. Specifically, we bypass the strong Gaussianity assumption that typically exists but fails in the unsupervised learning subtyping literature due to small-sized samples by vector quantization. Our proposed method better captures the latent space features and models the cancer subtype manifestation on a molecular basis, as demonstrated by the extensive experimental results.
Collapse
|
43
|
Machine learning in neuro-oncology: toward novel development fields. J Neurooncol 2022; 159:333-346. [PMID: 35761160 DOI: 10.1007/s11060-022-04068-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/11/2022] [Indexed: 10/17/2022]
Abstract
PURPOSE Artificial Intelligence (AI) involves several and different techniques able to elaborate a large amount of data responding to a specific planned outcome. There are several possible applications of this technology in neuro-oncology. METHODS We reviewed, according to PRISMA guidelines, available studies adopting AI in different fields of neuro-oncology including neuro-radiology, pathology, surgery, radiation therapy, and systemic treatments. RESULTS Neuro-radiology presented the major number of studies assessing AI. However, this technology is being successfully tested also in other operative settings including surgery and radiation therapy. In this context, AI shows to significantly reduce resources and costs maintaining an elevated qualitative standard. Pathological diagnosis and development of novel systemic treatments are other two fields in which AI showed promising preliminary data. CONCLUSION It is likely that AI will be quickly included in some aspects of daily clinical practice. Possible applications of these techniques are impressive and cover all aspects of neuro-oncology.
Collapse
|
44
|
Xu Z, Han Q, Yang D, Li Y, Shang Q, Liu J, Li W, Xu H, Chen Q. Automatic Detection of Image-Based Features for Immunosuppressive Therapy Response Prediction in Oral Lichen Planus. Front Immunol 2022; 13:942945. [PMID: 35812391 PMCID: PMC9260599 DOI: 10.3389/fimmu.2022.942945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Oral lichen planus (OLP) is a chronic inflammatory disease, and the common management focuses on controlling inflammation with immunosuppressive therapy. While the response to the immunosuppressive therapy is heterogeneous, exploring the mechanism and prediction of the response gain greater importance. Here, we developed a workflow for prediction of immunosuppressive therapy response prediction in OLP, which could automatically acquire image-based features. First, 38 features were acquired from 208 OLP pathological images, and 6 features were subsequently obtained which had a significant impact on the effect of OLP immunosuppressive therapy. By observing microscopic structure and integrated with the corresponding transcriptome, the biological implications of the 6 features were uncovered. Though the pathway enrichment analysis, three image-based features which advantageous to therapy indicated the different lymphocytes infiltration, and the other three image-based features which bad for therapy respectively indicated the nicotinamide adenine dinucleotide (NADH) metabolic pathway, response to potassium ion pathway and adenosine monophosphate (AMP) activated protein kinase pathway. In addition, prediction models for the response to immunosuppressive therapy, were constructed with above image-based features. The best performance prediction model built by logistic regression showed an accuracy of 90% and the area under the receiver operating characteristic curve (AUROC) reached 0.947. This study provided a novel approach to automatically obtain biological meaningful image-based features from unannotated pathological images, which could indicate the immunosuppressive therapy in OLP. Besides, the novel and accurate prediction model may be useful for the OLP clinical management.
Collapse
Affiliation(s)
- Ziang Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qi Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dan Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yijun Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianhui Shang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiaxin Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weiqi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hao Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Hao Xu, ; Qianming Chen,
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Affiliated Stomatology Hospital, Zhejiang University School of Stomatology, Hangzhou, China
- *Correspondence: Hao Xu, ; Qianming Chen,
| |
Collapse
|
45
|
Solary E, Abou-Zeid N, Calvo F. Ageing and cancer: a research gap to fill. Mol Oncol 2022; 16:3220-3237. [PMID: 35503718 PMCID: PMC9490141 DOI: 10.1002/1878-0261.13222] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/01/2022] [Accepted: 05/02/2022] [Indexed: 12/03/2022] Open
Abstract
The complex mechanisms of ageing biology are increasingly understood. Interventions to reduce or delay ageing‐associated diseases are emerging. Cancer is one of the diseases promoted by tissue ageing. A clockwise mutational signature is identified in many tumours. Ageing might be a modifiable cancer risk factor. To reduce the incidence of ageing‐related cancer and to detect the disease at earlier stages, we need to understand better the links between ageing and tumours. When a cancer is established, geriatric assessment and measures of biological age might help to generate evidence‐based therapeutic recommendations. In this approach, patients and caregivers would include the respective weight to give to the quality of life and survival in the therapeutic choices. The increasing burden of cancer in older patients requires new generations of researchers and geriatric oncologists to be trained, to properly address disease complexity in a multidisciplinary manner, and to reduce health inequities in this population of patients. In this review, we propose a series of research challenges to tackle in the next few years to better prevent, detect and treat cancer in older patients while preserving their quality of life.
Collapse
Affiliation(s)
- Eric Solary
- Fondation « Association pour la Recherche sur le Cancer », Villejuif, France.,Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France.,Gustave Roussy Cancer Center, INSERM U1287, Villejuif, France
| | - Nancy Abou-Zeid
- Fondation « Association pour la Recherche sur le Cancer », Villejuif, France
| | - Fabien Calvo
- Fondation « Association pour la Recherche sur le Cancer », Villejuif, France.,Université de Paris, Paris, France
| |
Collapse
|
46
|
Li M, Gong J, Bao Y, Huang D, Peng J, Tong T. Special issue "The advance of solid tumor research in China": Prognosis prediction for stage II colorectal cancer by fusing CT radiomics and deep-learning features of primary lesions and peripheral lymph nodes. Int J Cancer 2022; 152:31-41. [PMID: 35484979 DOI: 10.1002/ijc.34053] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/10/2022] [Accepted: 04/21/2022] [Indexed: 11/11/2022]
Abstract
Currently, the prognosis assessment of stage II colorectal cancer (CRC) remains a difficult clinical problem; therefore, more accurate prognostic predictors must be developed. In this study, we developed a prognostic prediction model for stage II CRC by fusing radiomics and deep-learning (DL) features of primary lesions and peripheral lymph nodes (LNs) in computed tomography (CT) scans. First, two CT radiomics models were built using primary lesion and LN image features. Subsequently, an information fusion method was used to build a fusion radiomics model by combining the tumor and LN image features. Furthermore, a transfer learning method was applied to build a deep convolutional neural network (CNN) model. Finally, the prediction scores generated by the radiomics and CNN models were fused to improve the prognosis prediction performance. The disease-free survival (DFS) and overall survival (OS) prediction areas under the curves (AUCs) generated by the fusion model improved to 0.76±0.08 and 0.91±0.05, respectively. These were significantly higher than the AUCs generated by the models using the individual CT radiomics and deep image features. Applying the survival analysis method, the DFS and OS fusion models yielded concordance index (C-index) values of 0.73 and 0.9, respectively. Hence, the combined model exhibited good predictive efficacy; therefore, it could be used for the accurate assessment of the prognosis of stage II CRC patients. Moreover, it could be used to screen out high-risk patients with poor prognoses, and assist in the formulation of clinical treatment decisions in a timely manner to achieve precision medicine. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Menglei Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P R China
| | - Jing Gong
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P R China
| | - Yichao Bao
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P R China
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P R China
| | - Junjie Peng
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P R China
| | - Tong Tong
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P R China
| |
Collapse
|
47
|
Generation of microbial colonies dataset with deep learning style transfer. Sci Rep 2022; 12:5212. [PMID: 35338253 PMCID: PMC8956727 DOI: 10.1038/s41598-022-09264-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/21/2022] [Indexed: 01/02/2023] Open
Abstract
We introduce an effective strategy to generate an annotated synthetic dataset of microbiological images of Petri dishes that can be used to train deep learning models in a fully supervised fashion. The developed generator employs traditional computer vision algorithms together with a neural style transfer method for data augmentation. We show that the method is able to synthesize a dataset of realistic looking images that can be used to train a neural network model capable of localising, segmenting, and classifying five different microbial species. Our method requires significantly fewer resources to obtain a useful dataset than collecting and labeling a whole large set of real images with annotations. We show that starting with only 100 real images, we can generate data to train a detector that achieves comparable results (detection mAP \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=0.416$$\end{document}=0.416, and counting MAE \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=4.49$$\end{document}=4.49) to the same detector but trained on a real, several dozen times bigger dataset (mAP \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=0.520$$\end{document}=0.520, MAE \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=4.31$$\end{document}=4.31), containing over 7 k images. We prove the usefulness of the method in microbe detection and segmentation, but we expect that it is general and flexible and can also be applicable in other domains of science and industry to detect various objects.
Collapse
|
48
|
King H, Wright J, Treanor D, Williams B, Randell R. What works where and how for uptake and impact of artificial intelligence in pathology: A review of theories for a realist evaluation (Preprint). J Med Internet Res 2022; 25:e38039. [PMID: 37093631 PMCID: PMC10167589 DOI: 10.2196/38039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/14/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND There is increasing interest in the use of artificial intelligence (AI) in pathology to increase accuracy and efficiency. To date, studies of clinicians' perceptions of AI have found only moderate acceptability, suggesting the need for further research regarding how to integrate it into clinical practice. OBJECTIVE The aim of the study was to determine contextual factors that may support or constrain the uptake of AI in pathology. METHODS To go beyond a simple listing of barriers and facilitators, we drew on the approach of realist evaluation and undertook a review of the literature to elicit stakeholders' theories of how, for whom, and in what circumstances AI can provide benefit in pathology. Searches were designed by an information specialist and peer-reviewed by a second information specialist. Searches were run on the arXiv.org repository, MEDLINE, and the Health Management Information Consortium, with additional searches undertaken on a range of websites to identify gray literature. In line with a realist approach, we also made use of relevant theory. Included documents were indexed in NVivo 12, using codes to capture different contexts, mechanisms, and outcomes that could affect the introduction of AI in pathology. Coded data were used to produce narrative summaries of each of the identified contexts, mechanisms, and outcomes, which were then translated into theories in the form of context-mechanism-outcome configurations. RESULTS A total of 101 relevant documents were identified. Our analysis indicates that the benefits that can be achieved will vary according to the size and nature of the pathology department's workload and the extent to which pathologists work collaboratively; the major perceived benefit for specialist centers is in reducing workload. For uptake of AI, pathologists' trust is essential. Existing theories suggest that if pathologists are able to "make sense" of AI, engage in the adoption process, receive support in adapting their work processes, and can identify potential benefits to its introduction, it is more likely to be accepted. CONCLUSIONS For uptake of AI in pathology, for all but the most simple quantitative tasks, measures will be required that either increase confidence in the system or provide users with an understanding of the performance of the system. For specialist centers, efforts should focus on reducing workload rather than increasing accuracy. Designers also need to give careful thought to usability and how AI is integrated into pathologists' workflow.
Collapse
Affiliation(s)
- Henry King
- Faculty of Medicine & Health, University of Leeds, Leeds, United Kingdom
| | - Judy Wright
- Faculty of Medicine & Health, University of Leeds, Leeds, United Kingdom
| | - Darren Treanor
- Faculty of Medicine & Health, University of Leeds, Leeds, United Kingdom
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
- Department of Clinical Pathology, and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization, Linköping University, Linköping, Sweden
| | | | - Rebecca Randell
- Faculty of Health Studies, University of Bradford, Bradford, United Kingdom
- Wolfson Centre for Applied Health Research, Bradford, United Kingdom
| |
Collapse
|
49
|
High-dimensional role of AI and machine learning in cancer research. Br J Cancer 2022; 126:523-532. [PMID: 35013580 PMCID: PMC8854697 DOI: 10.1038/s41416-021-01689-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 11/23/2021] [Accepted: 12/23/2021] [Indexed: 01/12/2023] Open
Abstract
The role of Artificial Intelligence and Machine Learning in cancer research offers several advantages, primarily scaling up the information processing and increasing the accuracy of the clinical decision-making. The key enabling tools currently in use in Precision, Digital and Translational Medicine, here named as 'Intelligent Systems' (IS), leverage unprecedented data volumes and aim to model their underlying heterogeneous influences and variables correlated with patients' outcomes. As functionality and performance of IS are associated with complex diagnosis and therapy decisions, a rich spectrum of patterns and features detected in high-dimensional data may be critical for inference purposes. Many challenges are also present in such discovery task. First, the generation of interpretable model results from a mix of structured and unstructured input information. Second, the design, and implementation of automated clinical decision processes for drawing disease trajectories and patient profiles. Ultimately, the clinical impacts depend on the data effectively subjected to steps such as harmonisation, integration, validation, etc. The aim of this work is to discuss the transformative value of IS applied to multimodal data acquired through various interrelated cancer domains (high-throughput genomics, experimental biology, medical image processing, radiomics, patient electronic records, etc.).
Collapse
|
50
|
Song J, Jiao W, Lankowicz K, Cai Z, Bi H. A two-stage adaptive thresholding segmentation for noisy low-contrast images. ECOL INFORM 2022. [DOI: 10.1016/j.ecoinf.2022.101632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|