1
|
Gupta A, Bajaj S, Nema P, Purohit A, Kashaw V, Soni V, Kashaw SK. Potential of AI and ML in oncology research including diagnosis, treatment and future directions: A comprehensive prospective. Comput Biol Med 2025; 189:109918. [PMID: 40037170 DOI: 10.1016/j.compbiomed.2025.109918] [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: 04/29/2024] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 03/06/2025]
Abstract
Artificial intelligence (AI) and machine learning (ML) have emerged as transformative tools in cancer research, offering the ability to process huge data rapidly and make precise therapeutic decisions. Over the last decade, AI, particularly deep learning (DL) and machine learning (ML), has significantly enhanced cancer prediction, diagnosis, and treatment by leveraging algorithms such as convolutional neural networks (CNNs) and multi-layer perceptrons (MLPs). These technologies provide reliable, efficient solutions for managing aggressive diseases like cancer, which have high recurrence and mortality rates. This review prospective highlights the applications of AI in oncology, a long with FDA-approved technologies like EFAI RTSuite CT HN-Segmentation System, Quantib Prostate, and Paige Prostate, and explore their role in advancing cancer detection, personalized care, and treatment. Furthermore, we also explored broader applications of AI in healthcare, addressing challenges, limitations, regulatory considerations, and ethical implications. By presenting these advancements, we underscore AI's potential to revolutionize cancer care, management and treatment.
Collapse
Affiliation(s)
- Akanksha Gupta
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, Madya Pradesh, 470003, India.
| | - Samyak Bajaj
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, Madya Pradesh, 470003, India.
| | - Priyanshu Nema
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, Madya Pradesh, 470003, India.
| | - Arpana Purohit
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, Madya Pradesh, 470003, India.
| | - Varsha Kashaw
- Sagar Institute of Pharmaceutical Sciences, Sagar, M.P., India.
| | - Vandana Soni
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, Madya Pradesh, 470003, India.
| | - Sushil K Kashaw
- Integrated Drug Discovery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, Madya Pradesh, 470003, India.
| |
Collapse
|
2
|
Mousavi R, Mustafa Ali MK, Lobo D. Discovery of Dynamic Models for AML Disease Progression from Longitudinal Multi-Modal Clinical Data Using Explainable Machine Learning. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.07.25325267. [PMID: 40297459 PMCID: PMC12036371 DOI: 10.1101/2025.04.07.25325267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Acute Myeloid Leukemia (AML) is a complex and heterogeneous disease identified by severe clinical progression, fast cellular proliferation, and often high mortality rates. Incorporating diverse longitudinal information on patients' medical histories is essential for developing effective disease predictive models applicable to both research and clinical settings. Here, we present a robust methodology for discovering dynamic predictive models to elucidate AML disease progression dynamics from a novel longitudinal multimodal clinical dataset of patients diagnosed with AML. The clinical dataset was analyzed to reveal the main clinical, genetic, and treatment features modulating disease progression. To discover mathematical models-including interactions, parameters, and nodes-predictive of AML progression, we present an explainable machine learning algorithm based on high-performance evolutionary computation. The results demonstrate that the predictive methodology could accurately estimate the clinical dynamics of AML progression in terms of blast percentages for both training and novel patients. This study demonstrates that the developed explainable machine learning approach can successfully predict AML progression by leveraging the heterogeneous and longitudinal dynamics of patients' clinical data. More importantly, this methodology shows significant potential for application in modeling the progression dynamics of other acute diseases, providing a flexible and adaptable framework for advancing clinical and translational research.
Collapse
Affiliation(s)
- Reza Mousavi
- Department of Biological Sciences, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Moaath K. Mustafa Ali
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Daniel Lobo
- Department of Biological Sciences, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center and Center for Stem Cell Biology & Regenerative Medicine, University of Maryland, School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, USA
| |
Collapse
|
3
|
Chan SPY, Rashid MBMA, Lim JJ, Goh JJN, Wong WY, Hooi L, Ismail NN, Luo B, Chen BJ, Noor NFBM, Phua BXM, Villanueva A, Sam XX, Ong CAJ, Chia CS, Abidin SZ, Yong MH, Kumar K, Ooi LL, Tay TKY, Woo XY, Toh TB, Yang VS, Chow EKH. Functional combinatorial precision medicine for predicting and optimizing soft tissue sarcoma treatments. NPJ Precis Oncol 2025; 9:83. [PMID: 40121334 PMCID: PMC11929909 DOI: 10.1038/s41698-025-00851-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/24/2025] [Indexed: 03/25/2025] Open
Abstract
Soft tissue sarcomas (STS) are rare, heterogeneous tumors with poor survival outcomes, primarily due to reliance on cytotoxic chemotherapy and lack of targeted therapies. Given the uniquely individualized nature of STS, we hypothesized that the ex vivo drug sensitivity platform, quadratic phenotypic optimization platform (QPOP), can predict treatment response and enhance combination therapy design for STS. Using QPOP, we screened 45 primary STS patient samples, and showed improved or concordant patient outcomes that are attributable to QPOP predictions. From a panel of approved and investigational agents, QPOP identified AZD5153 (BET inhibitor) and pazopanib (multi-kinase blocker) as the most effective combination with superior efficacy compared to standard regimens. Validation in a panel of established patient lines and in vivo models supported its synergistic interaction, accompanied by repressed oncogenic MYC and related pathways. These findings provide preliminary clinical evidence for QPOP to predict STS treatment outcomes and guide the development of novel therapeutic strategies for STS patients.
Collapse
Affiliation(s)
- Sharon Pei Yi Chan
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, #12-01 Centre for Translational Medicine, Singapore, 117599, Republic of Singapore
| | | | - Jhin Jieh Lim
- KYAN Technologies, 1 Research Link, #05-45, Singapore, 117604, Republic of Singapore
| | - Janice Jia Ni Goh
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Republic of Singapore
| | - Wai Yee Wong
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Republic of Singapore
| | - Lissa Hooi
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, #12-01 Centre for Translational Medicine, Singapore, 117599, Republic of Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, #12-01 Centre for Translational Medicine, Singapore, 117599, Republic of Singapore
| | - Nur Nadiah Ismail
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, 28 Medical Drive, #05-COR, Singapore, 117456, Republic of Singapore
| | - Baiwen Luo
- The N1 Institute for Health, National University of Singapore, 28 Medical Drive, Singapore, 117456, Republic of Singapore
| | - Benjamin Jieming Chen
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Nur Fazlin Bte Mohamed Noor
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore, 168583, Republic of Singapore
| | - Brandon Xuan Ming Phua
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Republic of Singapore
| | - Andre Villanueva
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Xin Xiu Sam
- Department of Anatomical Pathology, Singapore General Hospital, College Road, Level 7 Academia, Singapore, 169856, Republic of Singapore
| | - Chin-Ann Johnny Ong
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore, 168583, Republic of Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore, 168583, Republic of Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
| | - Claramae Shulyn Chia
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore, 168583, Republic of Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
| | - Suraya Zainul Abidin
- Department of Orthopaedic Surgery, Singapore General Hospital, 10 Hospital Boulevard, Tower Level 4 SingHealth Tower, Singapore, 168582, Republic of Singapore
| | - Ming-Hui Yong
- Department of Neurology, National Neuroscience Institute (Singapore General Hospital Campus), Outram Rd, Singapore, 169608, Republic of Singapore
| | - Krishan Kumar
- Department of Neurosurgery, National Neuroscience Institute (Singapore General Hospital Campus), Outram Rd, Singapore, 169608, Republic of Singapore
| | - London Lucien Ooi
- Oncology Academic Clinical Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
- Hepato-pancreato-biliary and Transplant Surgery, Singapore General Hospital, Outram Rd, Singapore, 169608, Republic of Singapore
| | - Timothy Kwang Yong Tay
- Department of Anatomical Pathology, Singapore General Hospital, College Road, Level 7 Academia, Singapore, 169856, Republic of Singapore
| | - Xing Yi Woo
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Republic of Singapore
| | - Tan Boon Toh
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, 28 Medical Drive, #05-COR, Singapore, 117456, Republic of Singapore.
- The N1 Institute for Health, National University of Singapore, 28 Medical Drive, Singapore, 117456, Republic of Singapore.
| | - Valerie Shiwen Yang
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore.
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore, 168583, Republic of Singapore.
- Oncology Academic Clinical Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore.
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, #12-01 Centre for Translational Medicine, Singapore, 117599, Republic of Singapore.
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, #12-01 Centre for Translational Medicine, Singapore, 117599, Republic of Singapore.
- The N1 Institute for Health, National University of Singapore, 28 Medical Drive, Singapore, 117456, Republic of Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Republic of Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 4 Engineering Drive 3, #04-08, Singapore, 117583, Republic of Singapore.
| |
Collapse
|
4
|
Jin X, Zhang J, Yang J, Yang S, Xue D, Zhang Z. Every cloud has a silver lining: DeepSeek's light through acute respiratory distress syndrome shadows. J Thorac Dis 2025; 17:1109-1113. [PMID: 40083514 PMCID: PMC11898352 DOI: 10.21037/jtd-2025-381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Affiliation(s)
- Xinhao Jin
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Zhang
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jie Yang
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Suibi Yang
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dong Xue
- The Key Laboratory of Smart Manufacturing in Energy Chemical Process, Ministry of Education, East China University of Science and Technology, Shanghai, China
| | - Zhongheng Zhang
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Shaoxing University, Shaoxing, China
- Longquan Industrial Innovation Research Institute, Lishui, China
| |
Collapse
|
5
|
Rao M, Luo W, Luo C, Wu B, Xu T, Wei Z, Deng H, Li K, Zhou D. Prognostic factors and outcomes in pediatric acute myeloid leukemia: a comprehensive bibliometric analysis of global research trends. Front Oncol 2025; 15:1466818. [PMID: 40034590 PMCID: PMC11873564 DOI: 10.3389/fonc.2025.1466818] [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: 07/26/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
Background Pediatric AML prognosis research has advanced significantly, yet gaps in understanding genetic and molecular interactions persist. Despite improved outcomes, relapse/refractory cases and personalized treatment integration remain critical clinical challenges. Objective To analyze the global research landscape on pediatric AML prognosis, highlight influential components and collaborations, and identify major potential research trends. Methods Publications on pediatric AML prognosis research from 1999 to 2023 were retrieved from the Clarivate Analytics Web of Science Core Collection (WoSCC) database. Bibliometric analysis was conducted using CiteSpace and VOSviewer to identify leading countries, prominent institutions, high-impact journals, key research categories, influential authors, and emerging research topics. Results The bibliometric analysis encompassed 924 publications, with St. Jude Children's Research Hospital emerging as the most prolific institution. The United States leads globally in terms of countries, institutions, journals, and authors. Todd A. Alonzo ranks highest in publication volume, while U. Creutzig leads in citations. The top research categories were Oncology, Hematology, and Pediatrics. Key research topics included genomics, transcriptomics, epigenomics, targeted therapies, immune therapy, and integrative diagnostic approaches. Conclusion This bibliometric analysis highlights significant advancements in pediatric AML prognosis over the past 25 years, driven by the integration of genetic markers, immunological insights, transcriptomics, and epigenomics, which have collectively transformed risk stratification and treatment strategies. Overcoming challenges, such as discovering new therapeutic targets and enhancing treatment combinations, will depend on global collaboration and advanced technologies to propel the field forward.
Collapse
Affiliation(s)
- Mingliang Rao
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenna Luo
- Department of Laboratory Medicine, Heyuan People’s Hospital, Heyuan, China
| | - Caiju Luo
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Baojing Wu
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tiantian Xu
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ziqian Wei
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haolan Deng
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kejing Li
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dunhua Zhou
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
Gill SS, Ponniah HS, Giersztein S, Anantharaj RM, Namireddy SR, Killilea J, Ramsay D, Salih A, Thavarajasingam A, Scurtu D, Jankovic D, Russo S, Kramer A, Thavarajasingam SG. The diagnostic and prognostic capability of artificial intelligence in spinal cord injury: A systematic review. BRAIN & SPINE 2025; 5:104208. [PMID: 40027293 PMCID: PMC11871462 DOI: 10.1016/j.bas.2025.104208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025]
Abstract
Background Artificial intelligence (AI) models have shown potential for diagnosing and prognosticating traumatic spinal cord injury (tSCI), but their clinical utility remains uncertain. Method ology: The primary aim was to evaluate the performance of AI algorithms in diagnosing and prognosticating tSCI. Subsequent systematic searching of seven databases identified studies evaluating AI models. PROBAST and TRIPOD tools were used to assess the quality and reporting of included studies (PROSPERO: CRD42023464722). Fourteen studies, comprising 20 models and 280,817 pooled imaging datasets, were included. Analysis was conducted in line with the SWiM guidelines. Results For prognostication, 11 studies predicted outcomes including AIS improvement (30%), mortality and ambulatory ability (20% each), and discharge or length of stay (10%). The mean AUC was 0.770 (range: 0.682-0.902), indicating moderate predictive performance. Diagnostic models utilising DTI, CT, and T2-weighted MRI with CNN-based segmentation achieved a weighted mean accuracy of 0.898 (range: 0.813-0.938), outperforming prognostic models. Conclusion AI demonstrates strong diagnostic accuracy (mean accuracy: 0.898) and moderate prognostic capability (mean AUC: 0.770) for tSCI. However, the lack of standardised frameworks and external validation limits clinical applicability. Future models should integrate multimodal data, including imaging, patient characteristics, and clinician judgment, to improve utility and alignment with clinical practice.
Collapse
Affiliation(s)
- Saran Singh Gill
- Imperial Brain & Spine Initiative, Imperial College London, London, United Kingdom
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Hariharan Subbiah Ponniah
- Imperial Brain & Spine Initiative, Imperial College London, London, United Kingdom
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Sho Giersztein
- Imperial Brain & Spine Initiative, Imperial College London, London, United Kingdom
| | | | - Srikar Reddy Namireddy
- Imperial Brain & Spine Initiative, Imperial College London, London, United Kingdom
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Joshua Killilea
- Imperial Brain & Spine Initiative, Imperial College London, London, United Kingdom
| | - DanieleS.C. Ramsay
- Imperial Brain & Spine Initiative, Imperial College London, London, United Kingdom
| | - Ahmed Salih
- Imperial Brain & Spine Initiative, Imperial College London, London, United Kingdom
| | | | - Daniel Scurtu
- Department of Neurosurgery, Universitätsmedizin Mainz, Mainz, Germany
| | - Dragan Jankovic
- Department of Neurosurgery, LMU University Hospital, LMU, Munich, Germany
| | - Salvatore Russo
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Andreas Kramer
- Department of Neurosurgery, LMU University Hospital, LMU, Munich, Germany
| | - Santhosh G. Thavarajasingam
- Imperial Brain & Spine Initiative, Imperial College London, London, United Kingdom
- Department of Neurosurgery, LMU University Hospital, LMU, Munich, Germany
| |
Collapse
|
7
|
Garuffo L, Leoni A, Gatta R, Bernardi S. The Applications of Machine Learning in the Management of Patients Undergoing Stem Cell Transplantation: Are We Ready? Cancers (Basel) 2025; 17:395. [PMID: 39941764 PMCID: PMC11816169 DOI: 10.3390/cancers17030395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/10/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a life-saving therapy for hematologic malignancies, such as leukemia and lymphoma and other severe conditions but is associated with significant risks, including graft versus host disease (GVHD), relapse, and treatment-related mortality. The increasing complexity of clinical, genomic, and biomarker data has spurred interest in machine learning (ML), which has emerged as a transformative tool to enhance decision-making and optimize outcomes in HSCT. This review examines the applications of ML in HSCT, focusing on donor selection, conditioning regimen, and prediction of post-transplant outcomes. Machine learning approaches, including decision trees, random forests, and neural networks, have demonstrated potential in improving donor compatibility algorithms, mortality and relapse prediction, and GVHD risk stratification. Integrating "omics" data with ML models has enabled the identification of novel biomarkers and the development of highly accurate predictive tools, supporting personalized treatment strategies. Despite promising advancements, challenges persist, including data standardization, algorithm interpretability, and ethical considerations regarding patient privacy. While ML holds promise for revolutionizing HSCT management, addressing these barriers through multicenter collaborations and regulatory frameworks remains essential for broader clinical adoption. In addition, the potential of ML can cope with some challenges such as data harmonization, patients' data protection, and availability of adequate infrastructure. Future research should prioritize larger datasets, multimodal data integration, and robust validation methods to fully realize ML's transformative potential in HSCT.
Collapse
Affiliation(s)
- Luca Garuffo
- Unit of Blood Disease and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili, 25123 Brescia, Italy; (L.G.); (S.B.)
- CREA (Centro di Ricerca Emato-Oncologica AIL), ASST Spedali Civili of Brescia, 25123 Brescia, Italy
| | - Alessandro Leoni
- Unit of Blood Disease and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili, 25123 Brescia, Italy; (L.G.); (S.B.)
- CREA (Centro di Ricerca Emato-Oncologica AIL), ASST Spedali Civili of Brescia, 25123 Brescia, Italy
| | - Roberto Gatta
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy;
| | - Simona Bernardi
- Unit of Blood Disease and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili, 25123 Brescia, Italy; (L.G.); (S.B.)
- CREA (Centro di Ricerca Emato-Oncologica AIL), ASST Spedali Civili of Brescia, 25123 Brescia, Italy
- National Center for Gene Therapy and Drugs Based on RNA Technology—CN3, 35122 Padua, Italy
| |
Collapse
|
8
|
Abdelaziz EH, Ismail R, Mabrouk MS, Amin E. Multi-omics data integration and analysis pipeline for precision medicine: Systematic review. Comput Biol Chem 2024; 113:108254. [PMID: 39447405 DOI: 10.1016/j.compbiolchem.2024.108254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/05/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Precision medicine has gained considerable popularity since the "one-size-fits-all" approach did not seem very effective or reflective of the complexity of the human body. Subsequently, since single-omics does not reflect the complexity of the human body's inner workings, it did not result in the expected advancement in the medical field. Therefore, the multi-omics approach has emerged. The multi-omics approach involves integrating data from different omics technologies, such as DNA sequencing, RNA sequencing, mass spectrometry, and others, using computational methods and then analyzing the integrated result for different downstream analysis applications such as survival analysis, cancer classification, or biomarker identification. Most of the recent reviews were constrained to discussing one aspect of the multi-omics analysis pipeline, such as the dimensionality reduction step, the integration methods, or the interpretability aspect; however, very few provide a comprehensive review of every step of the analysis. This study aims to give an overview of the multi-omics analysis pipeline, starting with the most popular multi-omics databases used in recent literature, dimensionality reduction techniques, details the different types of data integration techniques and their downstream analysis applications, describes the most commonly used evaluation metrics, highlights the importance of model interpretability, and lastly discusses the challenges and potential future work for multi-omics data integration in precision medicine.
Collapse
Affiliation(s)
| | - Rasha Ismail
- Faculty of Computer and Information Sciences, Ainshams University, Cairo, Egypt.
| | - Mai S Mabrouk
- Information Technology and Computer Science School, Nile University, Cairo, Egypt.
| | - Eman Amin
- Faculty of Computer and Information Sciences, Ainshams University, Cairo, Egypt.
| |
Collapse
|
9
|
Llaneza-Lago S, Fraser WD, Green D. Bayesian unsupervised clustering identifies clinically relevant osteosarcoma subtypes. Brief Bioinform 2024; 26:bbae665. [PMID: 39701601 DOI: 10.1093/bib/bbae665] [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: 09/06/2024] [Revised: 10/28/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024] Open
Abstract
Identification of cancer subtypes is a critical step for developing precision medicine. Most cancer subtyping is based on the analysis of RNA sequencing (RNA-seq) data from patient cohorts using unsupervised machine learning methods such as hierarchical cluster analysis, but these computational approaches disregard the heterogeneous composition of individual cancer samples. Here, we used a more sophisticated unsupervised Bayesian model termed latent process decomposition (LPD), which handles individual cancer sample heterogeneity and deconvolutes the structure of transcriptome data to provide clinically relevant information. The work was performed on the pediatric tumor osteosarcoma, which is a prototypical model for a rare and heterogeneous cancer. The LPD model detected three osteosarcoma subtypes. The subtype with the poorest prognosis was validated using independent patient datasets. This new stratification framework will be important for more accurate diagnostic labeling, expediting precision medicine, and improving clinical trial success. Our results emphasize the importance of using more sophisticated machine learning approaches (and for teaching deep learning and artificial intelligence) for RNA-seq data analysis, which may assist drug targeting and clinical management.
Collapse
Affiliation(s)
- Sergio Llaneza-Lago
- Biomedical Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - William D Fraser
- Bioanalytical Facility, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, United Kingdom
| | - Darrell Green
- Biomedical Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| |
Collapse
|
10
|
Chustecki M. Benefits and Risks of AI in Health Care: Narrative Review. Interact J Med Res 2024; 13:e53616. [PMID: 39556817 PMCID: PMC11612599 DOI: 10.2196/53616] [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: 10/12/2023] [Revised: 06/17/2024] [Accepted: 09/19/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND The integration of artificial intelligence (AI) into health care has the potential to transform the industry, but it also raises ethical, regulatory, and safety concerns. This review paper provides an in-depth examination of the benefits and risks associated with AI in health care, with a focus on issues like biases, transparency, data privacy, and safety. OBJECTIVE This study aims to evaluate the advantages and drawbacks of incorporating AI in health care. This assessment centers on the potential biases in AI algorithms, transparency challenges, data privacy issues, and safety risks in health care settings. METHODS Studies included in this review were selected based on their relevance to AI applications in health care, focusing on ethical, regulatory, and safety considerations. Inclusion criteria encompassed peer-reviewed articles, reviews, and relevant research papers published in English. Exclusion criteria included non-peer-reviewed articles, editorials, and studies not directly related to AI in health care. A comprehensive literature search was conducted across 8 databases: OVID MEDLINE, OVID Embase, OVID PsycINFO, EBSCO CINAHL Plus with Full Text, ProQuest Sociological Abstracts, ProQuest Philosopher's Index, ProQuest Advanced Technologies & Aerospace, and Wiley Cochrane Library. The search was last updated on June 23, 2023. Results were synthesized using qualitative methods to identify key themes and findings related to the benefits and risks of AI in health care. RESULTS The literature search yielded 8796 articles. After removing duplicates and applying the inclusion and exclusion criteria, 44 studies were included in the qualitative synthesis. This review highlights the significant promise that AI holds in health care, such as enhancing health care delivery by providing more accurate diagnoses, personalized treatment plans, and efficient resource allocation. However, persistent concerns remain, including biases ingrained in AI algorithms, a lack of transparency in decision-making, potential compromises of patient data privacy, and safety risks associated with AI implementation in clinical settings. CONCLUSIONS In conclusion, while AI presents the opportunity for a health care revolution, it is imperative to address the ethical, regulatory, and safety challenges linked to its integration. Proactive measures are required to ensure that AI technologies are developed and deployed responsibly, striking a balance between innovation and the safeguarding of patient well-being.
Collapse
Affiliation(s)
- Margaret Chustecki
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
11
|
Lee H, Ko N, Namgoong S, Ham S, Koo J. Recent advances in and applications of ex vivo drug sensitivity analysis for blood cancers. Blood Res 2024; 59:37. [PMID: 39503808 PMCID: PMC11541977 DOI: 10.1007/s44313-024-00032-8] [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: 07/31/2024] [Accepted: 09/06/2024] [Indexed: 11/09/2024] Open
Abstract
Blood cancers, including leukemia, multiple myeloma, and lymphoma, pose significant challenges owing to their heterogeneous nature and the limitations of traditional treatments. Precision medicine has emerged as a transformative approach that offers tailored therapeutic strategies based on individual patient profiles. Ex vivo drug sensitivity analysis is central to this advancement, which enables testing of patient-derived cancer cells against a panel of therapeutic agents to predict clinical responses. This review provides a comprehensive overview of the latest advancements in ex vivo drug sensitivity analyses and their application in blood cancers. We discuss the development of more comprehensive drug response metrics and the evaluation of drug combinations to identify synergistic interactions. Additionally, we present evaluation of the advanced therapeutics such as antibody-drug conjugates using ex vivo assays. This review describes the critical role of ex vivo drug sensitivity analyses in advancing precision medicine by examining technological innovations and clinical applications. Ultimately, these innovations are paving the way for more effective and individualized treatments, improving patient outcomes, and establishing new standards for the management of blood cancers.
Collapse
Affiliation(s)
- Haeryung Lee
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Nahee Ko
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Sujin Namgoong
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Seunghyok Ham
- ImpriMedKorea, Inc., Seoul, 03920, Republic of Korea
| | - Jamin Koo
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
- ImpriMedKorea, Inc., Seoul, 03920, Republic of Korea.
| |
Collapse
|
12
|
Sahib NRBM, Mohamed JS, Rashid MBMA, Jayalakshmi, Lin YC, Chee YL, Fan BE, De Mel S, Ooi MGM, Jen WY, Chow EKH. A Combinatorial Functional Precision Medicine Platform for Rapid Therapeutic Response Prediction in AML. Cancer Med 2024; 13:e70401. [PMID: 39560206 PMCID: PMC11574777 DOI: 10.1002/cam4.70401] [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: 08/14/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Despite advances made in targeted biomarker-based therapy for acute myeloid leukemia (AML) treatment, remission is often short and followed by relapse and acquired resistance. Functional precision medicine (FPM) efforts have been shown to improve therapy selection guidance by incorporating comprehensive biological data to tailor individual treatment. However, effectively managing complex biological data, while also ensuring rapid conversion of actionable insights into clinical utility remains challenging. METHODS We have evaluated the clinical applicability of quadratic phenotypic optimization platform (QPOP), to predict clinical response to combination therapies in AML and reveal patient-centric insights into combination therapy sensitivities. In this prospective study, 51 primary samples from newly diagnosed (ND) or refractory/relapsed (R/R) AML patients were evaluated by QPOP following ex vivo drug testing. RESULTS Individualized drug sensitivity reports were generated in 55/63 (87.3%) patient samples with a median turnaround time of 5 (4-10) days from sample collection to report generation. To evaluate clinical feasibility, QPOP-predicted response was compared to clinical treatment outcomes and indicated concordant results with 83.3% sensitivity and 90.9% specificity and an overall 86.2% accuracy. Serial QPOP analysis in a FLT3-mutant patient sample indicated decreased FLT3 inhibitor (FLT3i) sensitivity, which is concordant with increasing FLT3 allelic burden and drug resistance development. Forkhead box M1 (FOXM1)-AKT signaling was subsequently identified to contribute to resistance to FLT3i. CONCLUSION Overall, this study demonstrates the feasibility of applying QPOP as a functional combinatorial precision medicine platform to predict therapeutic sensitivities in AML and provides the basis for prospective clinical trials evaluating ex vivo-guided combination therapy.
Collapse
Affiliation(s)
- Noor Rashidha Binte Meera Sahib
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jameelah Sheik Mohamed
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | | | - Jayalakshmi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Yen Lin Chee
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Bingwen Eugene Fan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology, Tan Tock Seng Hospital, Singapore
- Lee Kong Chain School of Medicine, Nanyang Technological University, Singapore
- Department of Laboratory Medicine, Khoo Teck Puat Hospital, Singapore
| | - Sanjay De Mel
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Melissa Gaik Ming Ooi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Wei-Ying Jen
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore
| |
Collapse
|
13
|
Puri SS, Lath AK, Goel N, Admane PD, Garg P, Ethirajan R. Transformative Role of Artificial Intelligence in Reporting Haematology Cases: A Case Report. Cureus 2024; 16:e73274. [PMID: 39650924 PMCID: PMC11625413 DOI: 10.7759/cureus.73274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 12/11/2024] Open
Abstract
Artificial intelligence (AI) is transforming haematology reporting by improving accuracy, standardisation, and speed, addressing the need for timely and precise diagnostics. This study explores the use of the AI100 (SigTuple Technologies Private Limited, Bangalore, India) automated machine, a smart robotic microscope designed to automate the microscopic analysis of peripheral blood smears. Through the analysis of four haematology cases, this study demonstrates how AI technology facilitates efficient cell identification, enhances risk stratification, enables early detection of abnormalities, and accelerates diagnostic turnaround times. These advancements support pathologists in delivering improved patient care by augmenting traditional diagnostic methods. While AI can streamline processes and increase diagnostic accuracy, it is intended to complement, rather than replace, the expertise and judgement of skilled pathologists.
Collapse
Affiliation(s)
| | | | - Neha Goel
- Microbiology, GS Medical College and Hospital, Hapur, IND
| | | | - Pradeep Garg
- Surgery, GS Medical College and Hospital, Hapur, IND
| | - Renu Ethirajan
- Research and Development, SigTuple Technologies Private Limited, Bangalore, IND
| |
Collapse
|
14
|
Borek WE, Nobre L, Pedicona SF, Campbell AE, Christopher JA, Nawaz N, Perkins DN, Moreno-Cardoso P, Kelsall J, Ferguson HR, Patel B, Gallipoli P, Arruda A, Ambinder AJ, Thompson A, Williamson A, Ghiaur G, Minden MD, Gribben JG, Britton DJ, Cutillas PR, Dokal AD. Phosphoproteomics predict response to midostaurin plus chemotherapy in independent cohorts of FLT3-mutated acute myeloid leukaemia. EBioMedicine 2024; 108:105316. [PMID: 39293215 PMCID: PMC11424955 DOI: 10.1016/j.ebiom.2024.105316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Acute myeloid leukaemia (AML) is a bone marrow malignancy with poor prognosis. One of several treatments for AML is midostaurin combined with intensive chemotherapy (MIC), currently approved for FLT3 mutation-positive (FLT3-MP) AML. However, many patients carrying FLT3 mutations are refractory or experience an early relapse following MIC treatment, and might benefit more from receiving a different treatment. Development of a stratification method that outperforms FLT3 mutational status in predicting MIC response would thus benefit a large number of patients. METHODS We employed mass spectrometry phosphoproteomics to analyse 71 diagnosis samples of 47 patients with FLT3-MP AML who subsequently received MIC. We then used machine learning to identify biomarkers of response to MIC, and validated the resulting predictive model in two independent validation cohorts (n = 20). FINDINGS We identified three distinct phosphoproteomic AML subtypes amongst long-term survivors. The subtypes showed similar duration of MIC response, but different modulation of AML-implicated pathways, and exhibited distinct, highly-predictive biomarkers of MIC response. Using these biomarkers, we built a phosphoproteomics-based predictive model of MIC response, which we called MPhos. When applied to two retrospective real-world patient test cohorts (n = 20), MPhos predicted MIC response with 83% sensitivity and 100% specificity (log-rank p < 7∗10-5, HR = 0.005 [95% CI: 0-0.31]). INTERPRETATION In validation, MPhos outperformed the currently-used FLT3-based stratification method. Our findings have the potential to transform clinical decision-making, and highlight the important role that phosphoproteomics is destined to play in precision oncology. FUNDING This work was funded by Innovate UK grants (application numbers: 22217 and 10054602) and by Kinomica Ltd.
Collapse
Affiliation(s)
| | - Luis Nobre
- Kinomica Ltd, Alderley Park, Macclesfield, United Kingdom
| | | | - Amy E Campbell
- Kinomica Ltd, Alderley Park, Macclesfield, United Kingdom
| | | | - Nazrath Nawaz
- Kinomica Ltd, Alderley Park, Macclesfield, United Kingdom
| | | | | | - Janet Kelsall
- Kinomica Ltd, Alderley Park, Macclesfield, United Kingdom
| | | | - Bela Patel
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Paolo Gallipoli
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Andrea Arruda
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Alex J Ambinder
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, USA
| | | | | | - Gabriel Ghiaur
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, USA
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - John G Gribben
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | | - Pedro R Cutillas
- Kinomica Ltd, Alderley Park, Macclesfield, United Kingdom; Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Arran D Dokal
- Kinomica Ltd, Alderley Park, Macclesfield, United Kingdom.
| |
Collapse
|
15
|
Li Y, Yan F, Xiang J, Wang W, Xie K, Luo L. Identification and experimental validation of immune-related gene PPARG is involved in ulcerative colitis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167300. [PMID: 38880160 DOI: 10.1016/j.bbadis.2024.167300] [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: 02/23/2024] [Revised: 04/30/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND The pathophysiology of ulcerative colitis (UC) is believed to be heavily influenced by immunology, which presents challenges for both diagnosis and treatment. The main aims of this study are to deepen our understanding of the immunological characteristics associated with the disease and to identify valuable biomarkers for diagnosis and treatment. METHODS The UC datasets were sourced from the GEO database and were analyzed using unsupervised clustering to identify different subtypes of UC. Twelve machine learning algorithms and Deep learning model DNN were developed to identify potential UC biomarkers, with the LIME and SHAP methods used to explain the models' findings. PPI network is used to verify the identified key biomarkers, and then a network connecting super enhancers, transcription factors and genes is constructed. Single-cell sequencing technology was utilized to investigate the role of Peroxisome Proliferator Activated Receptor Gamma (PPARG) in UC and its correlation with macrophage infiltration. Furthermore, alterations in PPARG expression were validated through Western blot (WB) and immunohistochemistry (IHC) in both in vitro and in vivo experiments. RESULT By utilizing bioinformatics techniques, we were able to pinpoint PPARG as a key biomarker for UC. The expression of PPARG was significantly reduced in cell models, UC animal models, and colitis models induced by dextran sodium sulfate (DSS). Interestingly, overexpression of PPARG was able to restore intestinal barrier function in H2O2-induced IEC-6 cells. Additionally, immune-related differentially expressed genes (DEGs) allowed for efficient classification of UC samples into neutrophil and mitochondrial metabolic subtypes. A diagnostic model incorporating the three disease-specific genes PPARG, PLA2G2A, and IDO1 demonstrated high accuracy in distinguishing between the UC group and the control group. Furthermore, single-cell analysis revealed that decreased PPARG expression in colon tissue may contribute to the polarization of M1 macrophages through activation of inflammatory pathways. CONCLUSION In conclusion, PPARG, a gene related to immunity, has been established as a reliable potential biomarker for the diagnosis and treatment of UC. The immune response it controls plays a key role in the progression and development of UC by enabling interaction between characteristic biomarkers and immune infiltrating cells.
Collapse
Affiliation(s)
- Yang Li
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Fangfang Yan
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Jing Xiang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Wenjian Wang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Kangping Xie
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
16
|
Gutierrez JJG, Lau E, Dharmapalan S, Parker M, Chen Y, Álvarez MA, Wang D. Multi-output prediction of dose-response curves enables drug repositioning and biomarker discovery. NPJ Precis Oncol 2024; 8:209. [PMID: 39304771 DOI: 10.1038/s41698-024-00691-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Abstract
Drug response prediction is hampered by uncertainty in the measures of response and selection of doses. In this study, we propose a probabilistic multi-output model to simultaneously predict all dose-responses and uncover their biomarkers. By describing the relationship between genomic features and chemical properties to every response at every dose, our multi-output Gaussian Process (MOGP) models enable assessment of drug efficacy using any dose-response metric. This approach was tested across two drug screening studies and ten cancer types. Kullback-leibler divergence measured the importance of each feature and identified EZH2 gene as a novel biomarker of BRAF inhibitor response. We demonstrate the effectiveness of our MOGP models in accurately predicting dose-responses in different cancer types and when there is a limited number of drug screening experiments for training. Our findings highlight the potential of MOGP models in enhancing drug development pipelines by reducing data requirements and improving precision in dose-response predictions.
Collapse
Affiliation(s)
- Juan-José Giraldo Gutierrez
- National Heart and Lung Institute, Imperial College London, London, UK.
- Department of Computer Science, The University of Sheffield, Sheffield, UK.
| | - Evelyn Lau
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Subhashini Dharmapalan
- Department of Computer Science, The University of Sheffield, Sheffield, UK
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Melody Parker
- Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Yurui Chen
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Mathematics, National University of Singapore, Singapore, Republic of Singapore
| | - Mauricio A Álvarez
- Department of Computer Science, The University of Manchester, Manchester, UK
| | - Dennis Wang
- National Heart and Lung Institute, Imperial College London, London, UK.
- Department of Computer Science, The University of Sheffield, Sheffield, UK.
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore.
- Bioinformatics Institute (BII), Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore.
| |
Collapse
|
17
|
Khan M, Banerjee S, Muskawad S, Maity R, Chowdhury SR, Ejaz R, Kuuzie E, Satnarine T. The Impact of Artificial Intelligence on Allergy Diagnosis and Treatment. Curr Allergy Asthma Rep 2024; 24:361-372. [PMID: 38954325 DOI: 10.1007/s11882-024-01152-y] [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] [Accepted: 05/19/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW Artificial intelligence (AI), be it neuronal networks, machine learning or deep learning, has numerous beneficial effects on healthcare systems; however, its potential applications and diagnostic capabilities for immunologic diseases have yet to be explored. Understanding AI systems can help healthcare workers better assimilate artificial intelligence into their practice and unravel its potential in diagnostics, clinical research, and disease management. RECENT FINDINGS We reviewed recent advancements in AI systems and their integration in healthcare systems, along with their potential benefits in the diagnosis and management of diseases. We explored machine learning as employed in allergy diagnosis and its learning patterns from patient datasets, as well as the possible advantages of using AI in the field of research related to allergic reactions and even remote monitoring. Considering the ethical challenges and privacy concerns raised by clinicians and patients with regard to integrating AI in healthcare, we explored the new guidelines adapted by regulatory bodies. Despite these challenges, AI appears to have been successfully incorporated into various healthcare systems and is providing patient-centered solutions while simultaneously assisting healthcare workers. Artificial intelligence offers new hope in the field of immunologic disease diagnosis, monitoring, and management and thus has the potential to revolutionize healthcare systems.
Collapse
Affiliation(s)
- Maham Khan
- Fatima Jinnah Medical University, Lahore, Pakistan.
| | | | | | - Rick Maity
- Institute of Post Graduate Medical Education and Research, Kolkata, West Bengal, India
| | | | - Rida Ejaz
- Shifa College of Medicine, Islamabad, Pakistan
| | | | | |
Collapse
|
18
|
Carvalho MFL, de Almeida BO, Bueno MLP, Vicari HP, Lima K, Rego EM, Roversi FM, Machado-Neto JA. Comprehensive analysis of the HCK gene in myeloid neoplasms: Insights into biological functions, prognosis, and response to antineoplastic agents. Hematol Transfus Cell Ther 2024; 46:273-282. [PMID: 38326180 PMCID: PMC11221266 DOI: 10.1016/j.htct.2023.11.007] [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: 08/19/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 02/09/2024] Open
Abstract
Myeloid neoplasms result from molecular alterations in hematopoietic stem cells, with acute myeloid leukemia (AML) being one of the most aggressive and with a poor prognosis. Hematopoietic cell kinase (HCK) is a proto-oncogene that encodes a protein-tyrosine kinase of the Scr family, and it is highly expressed in AML. The present study investigated HCK expression in normal hematopoietic cells across myeloid differentiation stages and myeloid neoplasm patients. Within the AML cohort, we explored the impact of HCK expression on clinical outcomes and its correlation with clinical, genetic, and laboratory characteristics. Furthermore, we evaluated the association between HCK expression and the response to antineoplastic agents using ex vivo assay data from AML patients. HCK expression is higher in differentiated subpopulations of myeloid cells. High HCK expression was observed in patients with chronic myelomonocytic leukemia, chronic myeloid leukemia, and AML. In patients with AML, high levels of HCK negatively impacted overall and disease-free survival. High HCK expression was also associated with worse molecular risk groups and white blood cell count; however, it was not an independent prognostic factor. In functional genomic analyses, high HCK expression was associated with several biological and molecular processes relevant to leukemogenesis. HCK expression was also associated with sensitivity and resistance to several drugs currently used in the clinic. In conclusion, our analysis confirmed the differential expression of HCK in myeloid neoplasms and its potential association with unfavorable molecular risks in AML. We also provide new insights into HCK biological functions, prognosis, and response to antineoplastic agents.
Collapse
Affiliation(s)
| | | | - Maura Lima Pereira Bueno
- Hematology and Transfusion Medicine Center, University of Campinas, Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | - Hugo Passos Vicari
- Institute of Biomedical Sciences, University of São Paulo (USP), SP, Brazil
| | - Keli Lima
- Institute of Biomedical Sciences, University of São Paulo (USP), SP, Brazil; Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Eduardo Magalhães Rego
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Fernanda Marconi Roversi
- Hematology and Transfusion Medicine Center, University of Campinas, Hemocentro-UNICAMP, Campinas, São Paulo, Brazil; Department of Surgery Division Emory University, Atlanta, GA, USA
| | | |
Collapse
|
19
|
Wei W, Fang J, Yang N, Li Q, Hu L, Zhao L, Han J. AC-ModNet: Molecular Reverse Design Network Based on Attribute Classification. Int J Mol Sci 2024; 25:6940. [PMID: 39000049 PMCID: PMC11241775 DOI: 10.3390/ijms25136940] [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: 05/12/2024] [Revised: 06/13/2024] [Accepted: 06/22/2024] [Indexed: 07/14/2024] Open
Abstract
Deep generative models are becoming a tool of choice for exploring the molecular space. One important application area of deep generative models is the reverse design of drug compounds for given attributes (solubility, ease of synthesis, etc.). Although there are many generative models, these models cannot generate specific intervals of attributes. This paper proposes a AC-ModNet model that effectively combines VAE with AC-GAN to generate molecular structures in specific attribute intervals. The AC-ModNet is trained and evaluated using the open 250K ZINC dataset. In comparison with related models, our method performs best in the FCD and Frag model evaluation indicators. Moreover, we prove the AC-ModNet created molecules have potential application value in drug design by comparing and analyzing them with medical records in the PubChem database. The results of this paper will provide a new method for machine learning drug reverse design.
Collapse
Affiliation(s)
| | | | - Ning Yang
- School of Automation, Northwestern Polytechnical University, Xi’an 710072, China; (W.W.); (J.F.); (Q.L.); (L.H.); (L.Z.); (J.H.)
| | | | | | | | | |
Collapse
|
20
|
Qin G, Dai J, Chien S, Martins TJ, Loera B, Nguyen QH, Oakes ML, Tercan B, Aguilar B, Hagen L, McCune J, Gelinas R, Monnat RJ, Shmulevich I, Becker PS. Mutation Patterns Predict Drug Sensitivity in Acute Myeloid Leukemia. Clin Cancer Res 2024; 30:2659-2671. [PMID: 38619278 PMCID: PMC11176916 DOI: 10.1158/1078-0432.ccr-23-1674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/15/2023] [Accepted: 12/08/2023] [Indexed: 04/16/2024]
Abstract
PURPOSE The inherent genetic heterogeneity of acute myeloid leukemia (AML) has challenged the development of precise and effective therapies. The objective of this study was to elucidate the genomic basis of drug resistance or sensitivity, identify signatures for drug response prediction, and provide resources to the research community. EXPERIMENTAL DESIGN We performed targeted sequencing, high-throughput drug screening, and single-cell genomic profiling on leukemia cell samples derived from patients with AML. Statistical approaches and machine learning models were applied to identify signatures for drug response prediction. We also integrated large public datasets to understand the co-occurring mutation patterns and further investigated the mutation profiles in the single cells. The features revealed in the co-occurring or mutual exclusivity pattern were further subjected to machine learning models. RESULTS We detected genetic signatures associated with sensitivity or resistance to specific agents, and identified five co-occurring mutation groups. The application of single-cell genomic sequencing unveiled the co-occurrence of variants at the individual cell level, highlighting the presence of distinct subclones within patients with AML. Using the mutation pattern for drug response prediction demonstrates high accuracy in predicting sensitivity to some drug classes, such as MEK inhibitors for RAS-mutated leukemia. CONCLUSIONS Our study highlights the importance of considering the gene mutation patterns for the prediction of drug response in AML. It provides a framework for categorizing patients with AML by mutations that enable drug sensitivity prediction.
Collapse
Affiliation(s)
| | - Jin Dai
- Division of Hematology, University of Washington, Seattle, Washington
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| | - Sylvia Chien
- Division of Hematology, University of Washington, Seattle, Washington
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| | - Timothy J. Martins
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| | - Brenda Loera
- City of Hope National Medical Center, Duarte, California
| | - Quy H. Nguyen
- University of California, Irvine, Irvine, California
| | | | - Bahar Tercan
- Institute for Systems Biology, Seattle, Washington
| | | | - Lauren Hagen
- Institute for Systems Biology, Seattle, Washington
| | | | | | - Raymond J. Monnat
- Lab Medicine|Pathology and Genome Sciences, University of Washington, Seattle, Washington
| | | | - Pamela S. Becker
- Division of Hematology, University of Washington, Seattle, Washington
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
- City of Hope National Medical Center, Duarte, California
| |
Collapse
|
21
|
Lingam G, Shakir T, Kader R, Chand M. Role of artificial intelligence in colorectal cancer. Artif Intell Gastrointest Endosc 2024; 5:90723. [DOI: 10.37126/aige.v5.i2.90723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/10/2024] [Accepted: 04/19/2024] [Indexed: 05/11/2024] Open
Abstract
The sphere of artificial intelligence (AI) is ever expanding. Applications for clinical practice have been emerging over recent years. Although its uptake has been most prominent in endoscopy, this represents only one aspect of holistic patient care. There are a multitude of other potential avenues in which gastrointestinal care may be involved. We aim to review the role of AI in colorectal cancer as a whole. We performed broad scoping and focused searches of the applications of AI in the field of colorectal cancer. All trials including qualitative research were included from the year 2000 onwards. Studies were grouped into pre-operative, intra-operative and post-operative aspects. Pre-operatively, the major use is with endoscopic recognition. Colonoscopy has embraced the use for human derived classifications such as Narrow-band Imaging International Colorectal Endoscopic, Japan Narrow-band Imaging Expert Team, Paris and Kudo. However, novel detection and diagnostic methods have arisen from advances in AI classification. Intra-operatively, adjuncts such as image enhanced identification of structures and assessment of perfusion have led to improvements in clinical outcomes. Post-operatively, monitoring and surveillance have taken strides with potential socioeconomic and environmental savings. The uses of AI within the umbrella of colorectal surgery are multiple. We have identified existing technologies which are already augmenting cancer care. The future applications are exciting and could at least match, if not surpass human standards.
Collapse
Affiliation(s)
- Gita Lingam
- Department of General Surgery, Princess Alexandra Hospital, Harlow CM20 1QX, United Kingdom
| | - Taner Shakir
- Department of Colorectal Surgery, University College London, London W1W 7TY, United Kingdom
| | - Rawen Kader
- Department of Gastroenterology, University College London, University College London Hospitals Nhs Foundation Trust, London W1B, United Kingdom
| | - Manish Chand
- Gastroenterological Intervention Centre, University College London, London W1W 7TS, United Kingdom
| |
Collapse
|
22
|
Shao W, Wang Y, Liu L, Ren Y, Wang J, Cui Y, Liu J, Zhang X, Zhang S, Liu S, Jiang E, Feng S, Pei X. Combining serum microRNAs and machine learning algorithms for diagnosing infectious fever after HSCT. Ann Hematol 2024; 103:2089-2102. [PMID: 38691145 DOI: 10.1007/s00277-024-05755-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/11/2024] [Indexed: 05/03/2024]
Abstract
Infection post-hematopoietic stem cell transplantation (HSCT) is one of the main causes of patient mortality. Fever is the most crucial clinical symptom indicating infection. However, current microbial detection methods are limited. Therefore, timely diagnosis of infectious fever and administration of antimicrobial drugs can effectively reduce patient mortality. In this study, serum samples were collected from 181 patients with HSCT with or without infection, as well as the clinical information. And more than 80 infectious-related microRNAs in the serum were selected according to the bulk RNA-seq result and detected in the 345 time-pointed serum samples by Q-PCR. Unsupervised clustering result indicates a close association between these microRNAs expression and infection occurrence. Compared to the uninfected cohort, more than 10 serum microRNAs were identified as the combined diagnostic markers in one formula constructed by the Random Forest (RF) algorithms, with a diagnostic accuracy more than 0.90. Furthermore, correlations of serum microRNAs to immune cells, inflammatory factors, pathgens, infection tissue, and prognosis were analyzed in the infection cohort. Overall, this study demonstrates that the combination of serum microRNAs detection and machine learning algorithms holds promising potential in diagnosing infectious fever after HSCT.
Collapse
Affiliation(s)
- Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Yixuan Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Li Liu
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yiran Ren
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Jieru Wang
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yuqing Cui
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Jia Liu
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Xiaoyu Zhang
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Sudong Zhang
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Shuangjie Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
| | - Xiaolei Pei
- State Key Laboratory of Experimental Hematology, Hematopoietic Stem Cell Transplantation Center, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
| |
Collapse
|
23
|
Karabacak M, Bhimani AD, Schupper AJ, Carr MT, Steinberger J, Margetis K. Machine learning models on a web application to predict short-term postoperative outcomes following anterior cervical discectomy and fusion. BMC Musculoskelet Disord 2024; 25:401. [PMID: 38773464 PMCID: PMC11110429 DOI: 10.1186/s12891-024-07528-5] [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: 01/17/2024] [Accepted: 05/15/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND The frequency of anterior cervical discectomy and fusion (ACDF) has increased up to 400% since 2011, underscoring the need to preoperatively anticipate adverse postoperative outcomes given the procedure's expanding use. Our study aims to accomplish two goals: firstly, to develop a suite of explainable machine learning (ML) models capable of predicting adverse postoperative outcomes following ACDF surgery, and secondly, to embed these models in a user-friendly web application, demonstrating their potential utility. METHODS We utilized data from the National Surgical Quality Improvement Program database to identify patients who underwent ACDF surgery. The outcomes of interest were four short-term postoperative adverse outcomes: prolonged length of stay (LOS), non-home discharges, 30-day readmissions, and major complications. We utilized five ML algorithms - TabPFN, TabNET, XGBoost, LightGBM, and Random Forest - coupled with the Optuna optimization library for hyperparameter tuning. To bolster the interpretability of our models, we employed SHapley Additive exPlanations (SHAP) for evaluating predictor variables' relative importance and used partial dependence plots to illustrate the impact of individual variables on the predictions generated by our top-performing models. We visualized model performance using receiver operating characteristic (ROC) curves and precision-recall curves (PRC). Quantitative metrics calculated were the area under the ROC curve (AUROC), balanced accuracy, weighted area under the PRC (AUPRC), weighted precision, and weighted recall. Models with the highest AUROC values were selected for inclusion in a web application. RESULTS The analysis included 57,760 patients for prolonged LOS [11.1% with prolonged LOS], 57,780 for non-home discharges [3.3% non-home discharges], 57,790 for 30-day readmissions [2.9% readmitted], and 57,800 for major complications [1.4% with major complications]. The top-performing models, which were the ones built with the Random Forest algorithm, yielded mean AUROCs of 0.776, 0.846, 0.775, and 0.747 for predicting prolonged LOS, non-home discharges, readmissions, and complications, respectively. CONCLUSIONS Our study employs advanced ML methodologies to enhance the prediction of adverse postoperative outcomes following ACDF. We designed an accessible web application to integrate these models into clinical practice. Our findings affirm that ML tools serve as vital supplements in risk stratification, facilitating the prediction of diverse outcomes and enhancing patient counseling for ACDF.
Collapse
Affiliation(s)
- Mert Karabacak
- Department of Neurosurgery, Mount Sinai Health System, 1468 Madison Ave, New York, NY, 10029, USA
| | - Abhiraj D Bhimani
- Department of Neurosurgery, Mount Sinai Health System, 1468 Madison Ave, New York, NY, 10029, USA
| | - Alexander J Schupper
- Department of Neurosurgery, Mount Sinai Health System, 1468 Madison Ave, New York, NY, 10029, USA
| | - Matthew T Carr
- Department of Neurosurgery, Mount Sinai Health System, 1468 Madison Ave, New York, NY, 10029, USA
| | - Jeremy Steinberger
- Department of Neurosurgery, Mount Sinai Health System, 1468 Madison Ave, New York, NY, 10029, USA
| | - Konstantinos Margetis
- Department of Neurosurgery, Mount Sinai Health System, 1468 Madison Ave, New York, NY, 10029, USA.
| |
Collapse
|
24
|
Johnson R, Stephens AV, Mester R, Knyazev S, Kohn LA, Freund MK, Bondhus L, Hill BL, Schwarz T, Zaitlen N, Arboleda VA, Bastarache LA, Pasaniuc B, Butte MJ. Electronic health record signatures identify undiagnosed patients with common variable immunodeficiency disease. Sci Transl Med 2024; 16:eade4510. [PMID: 38691621 PMCID: PMC11402387 DOI: 10.1126/scitranslmed.ade4510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 04/10/2024] [Indexed: 05/03/2024]
Abstract
Human inborn errors of immunity include rare disorders entailing functional and quantitative antibody deficiencies due to impaired B cells called the common variable immunodeficiency (CVID) phenotype. Patients with CVID face delayed diagnoses and treatments for 5 to 15 years after symptom onset because the disorders are rare (prevalence of ~1/25,000), and there is extensive heterogeneity in CVID phenotypes, ranging from infections to autoimmunity to inflammatory conditions, overlapping with other more common disorders. The prolonged diagnostic odyssey drives excessive system-wide costs before diagnosis. Because there is no single causal mechanism, there are no genetic tests to definitively diagnose CVID. Here, we present PheNet, a machine learning algorithm that identifies patients with CVID from their electronic health records (EHRs). PheNet learns phenotypic patterns from verified CVID cases and uses this knowledge to rank patients by likelihood of having CVID. PheNet could have diagnosed more than half of our patients with CVID 1 or more years earlier than they had been diagnosed. When applied to a large EHR dataset, followed by blinded chart review of the top 100 patients ranked by PheNet, we found that 74% were highly probable to have CVID. We externally validated PheNet using >6 million records from disparate medical systems in California and Tennessee. As artificial intelligence and machine learning make their way into health care, we show that algorithms such as PheNet can offer clinical benefits by expediting the diagnosis of rare diseases.
Collapse
Affiliation(s)
- Ruth Johnson
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Alexis V. Stephens
- Department of Pediatrics, Division of Immunology, Allergy and Rheumatology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Rachel Mester
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sergey Knyazev
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Lisa A. Kohn
- Department of Pediatrics, Division of Immunology, Allergy and Rheumatology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Malika K. Freund
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Leroy Bondhus
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Brian L. Hill
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Tommer Schwarz
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Noah Zaitlen
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Valerie A. Arboleda
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Computational Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Lisa A. Bastarache
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA 37203
| | - Bogdan Pasaniuc
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Computational Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Manish J. Butte
- Department of Pediatrics, Division of Immunology, Allergy and Rheumatology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
25
|
Zhang B, Liu H, Wu F, Ding Y, Wu J, Lu L, Bajpai AK, Sang M, Wang X. Identification of hub genes and potential molecular mechanisms related to drug sensitivity in acute myeloid leukemia based on machine learning. Front Pharmacol 2024; 15:1359832. [PMID: 38650628 PMCID: PMC11033397 DOI: 10.3389/fphar.2024.1359832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
Abstract
Background: Acute myeloid leukemia (AML) is the most common form of leukemia among adults and is characterized by uncontrolled proliferation and clonal expansion of hematopoietic cells. There has been a significant improvement in the treatment of younger patients, however, prognosis in the elderly AML patients remains poor. Methods: We used computational methods and machine learning (ML) techniques to identify and explore the differential high-risk genes (DHRGs) in AML. The DHRGs were explored through multiple in silico approaches including genomic and functional analysis, survival analysis, immune infiltration, miRNA co-expression and stemness features analyses to reveal their prognostic importance in AML. Furthermore, using different ML algorithms, prognostic models were constructed and validated using the DHRGs. At the end molecular docking studies were performed to identify potential drug candidates targeting the selected DHRGs. Results: We identified a total of 80 DHRGs by comparing the differentially expressed genes derived between AML patients and normal controls and high-risk AML genes identified by Cox regression. Genetic and epigenetic alteration analyses of the DHRGs revealed a significant association of their copy number variations and methylation status with overall survival (OS) of AML patients. Out of the 137 models constructed using different ML algorithms, the combination of Ridge and plsRcox maintained the highest mean C-index and was used to build the final model. When AML patients were classified into low- and high-risk groups based on DHRGs, the low-risk group had significantly longer OS in the AML training and validation cohorts. Furthermore, immune infiltration, miRNA coexpression, stemness feature and hallmark pathway analyses revealed significant differences in the prognosis of the low- and high-risk AML groups. Drug sensitivity and molecular docking studies revealed top 5 drugs, including carboplatin and austocystin-D that may significantly affect the DHRGs in AML. Conclusion: The findings from the current study identified a set of high-risk genes that may be used as prognostic and therapeutic markers for AML patients. In addition, significant use of the ML algorithms in constructing and validating the prognostic models in AML was demonstrated. Although our study used extensive bioinformatics and machine learning methods to identify the hub genes in AML, their experimental validations using knock-out/-in methods would strengthen our findings.
Collapse
Affiliation(s)
- Boyu Zhang
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Haiyan Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Fengxia Wu
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yuhong Ding
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Jiarun Wu
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Lu Lu
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Akhilesh K. Bajpai
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Mengmeng Sang
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Xinfeng Wang
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
26
|
Li X, Xu H, Du Z, Cao Q, Liu X. Advances in the study of tertiary lymphoid structures in the immunotherapy of breast cancer. Front Oncol 2024; 14:1382701. [PMID: 38628669 PMCID: PMC11018917 DOI: 10.3389/fonc.2024.1382701] [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: 02/13/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Breast cancer, as one of the most common malignancies in women, exhibits complex and heterogeneous pathological characteristics across different subtypes. Triple-negative breast cancer (TNBC) and HER2-positive breast cancer are two common and highly invasive subtypes within breast cancer. The stability of the breast microbiota is closely intertwined with the immune environment, and immunotherapy is a common approach for treating breast cancer.Tertiary lymphoid structures (TLSs), recently discovered immune cell aggregates surrounding breast cancer, resemble secondary lymphoid organs (SLOs) and are associated with the prognosis and survival of some breast cancer patients, offering new avenues for immunotherapy. Machine learning, as a form of artificial intelligence, has increasingly been used for detecting biomarkers and constructing tumor prognosis models. This article systematically reviews the latest research progress on TLSs in breast cancer and the application of machine learning in the detection of TLSs and the study of breast cancer prognosis. The insights provided contribute valuable perspectives for further exploring the biological differences among different subtypes of breast cancer and formulating personalized treatment strategies.
Collapse
Affiliation(s)
- Xin Li
- The First Clinical School of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Han Xu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ziwei Du
- The First Clinical School of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiang Cao
- Department of Earth Sciences, Kunming University of Science and Technology, Kunming, China
| | - Xiaofei Liu
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
27
|
Romagnoli A, Ferrara F, Langella R, Zovi A. Healthcare Systems and Artificial Intelligence: Focus on Challenges and the International Regulatory Framework. Pharm Res 2024; 41:721-730. [PMID: 38443632 DOI: 10.1007/s11095-024-03685-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/28/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Nowadays, healthcare systems are coping with the challenge of countering the exponential growth of healthcare costs worldwide, to support sustainability and to guarantee access to treatment for all patients. METHODS Artificial Intelligence (AI) is the technology able to perform human cognitive functions through the creation of algorithms. The value of AI in healthcare and its ability to address healthcare delivery issues has been a subject of discussion within the scientific community for several years. RESULTS The aim of this work is to provide an overview of the primary uses of AI in the healthcare system, to discuss its desirable future uses while shedding light on the major issues related to implications within international regulatory processes. In this manuscript, it will be described the main applications of AI in various aspects of health care, from clinical studies to ethical implications, focusing on the international regulatory framework in countries in which AI is used, to discuss and compare strengthens and weaknesses. CONCLUSIONS The challenges in regulatory processes to facilitate the integration of AI in healthcare are significant. However, overcoming them is essential to ensure that AI-based technologies are adopted safely and effectively.
Collapse
Affiliation(s)
- Alessia Romagnoli
- Territorial Pharmaceutical Service, Local Health Unit Lanciano Vasto Chieti, Chieti, Italy
| | - Francesco Ferrara
- Pharmaceutical Department, Asl Napoli 3 Sud, Dell'amicizia street 22, 80035, Nola, Naples, Italy.
| | - Roberto Langella
- Italian Society of Hospital Pharmacy (SIFO), SIFO Secretariat of the Lombardy Region, Carlo Farini street, 81, 20159, Milan, Italy
| | - Andrea Zovi
- Ministry of Health, Viale Giorgio Ribotta 5, 00144, Rome, Italy
| |
Collapse
|
28
|
Park S, Kim TY, Cho BS, Kwag D, Lee JM, Kim M, Kim Y, Koo J, Raman A, Kim TK, Kim HJ. Prognostic value of European LeukemiaNet 2022 criteria and genomic clusters using machine learning in older adults with acute myeloid leukemia. Haematologica 2024; 109:1095-1106. [PMID: 37706344 PMCID: PMC10985444 DOI: 10.3324/haematol.2023.283606] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023] Open
Abstract
This study aimed to validate the new European Leukemia Net (ELN) 2022 criteria for genetic risk stratification in older adults with acute myeloid leukemia (AML) and to determine the most likely set of clusters of similar cytogenetic and mutation properties correlated with survival outcomes in three treatment groups: intensive chemotherapy (IC), hypomethylating agents (HMA) alone, and HMA plus venetoclax (HMA/VEN). The study included 279 patients (aged ≥60 years) who received IC (N=131), HMA (N=76), and HMA/VEN (N=72) between July 2017 and October 2021. No significant differences were observed in survival among the groups according to ELN 2022 risk stratification. Unsupervised hierarchical clustering analysis identified nine genomic clusters (C1-9) with varying survival outcomes depending on treatment type. For example, C4 (predominant for core binding factor-AML) displayed a favorable prognosis in the IC group, but not in the HMA or HMA/VEN groups. The HMA/VEN group had better outcomes than the HMA group in many clusters (C1, 2, 3, and 5); however, the addition of VEN to HMA or IC did not improve the survival outcomes compared with those of HMA alone in C7 and C9 (predominant for -5, del(5q), -7, -17/abn(17p), complex karyotypes, and mutated TP53). The study highlights the limitations of ELN genetic risk stratification in older adults with AML. It emphasizes the need for a more comprehensive approach that considers co-occurring somatic mutations to guide treatment selection in older adults with AML.
Collapse
Affiliation(s)
- Silvia Park
- Department of Hematology, Catholic Hematology Hospital, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea; Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea; Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Tong Yoon Kim
- Department of Hematology, Catholic Hematology Hospital, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea; Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul
| | - Byung-Sik Cho
- Department of Hematology, Catholic Hematology Hospital, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea; Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul.
| | - Daehun Kwag
- Department of Hematology, Catholic Hematology Hospital, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea; Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul
| | - Jong-Mi Lee
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul
| | - MyungShin Kim
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul
| | - Yonggoo Kim
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul
| | - Jamin Koo
- Department of Chemical Engineering, Hongik University, Seoul, Korea; ImpriMedKorea Inc, Seoul
| | - Anjali Raman
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University, Nashville, TN
| | - Tae Kon Kim
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University, Nashville, TN
| | - Hee-Je Kim
- Department of Hematology, Catholic Hematology Hospital, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea; Leukemia Research Institute, College of Medicine, The Catholic University of Korea, Seoul
| |
Collapse
|
29
|
Al-Hussaini I, White B, Varmeziar A, Mehra N, Sanchez M, Lee J, DeGroote NP, Miller TP, Mitchell CS. An Interpretable Machine Learning Framework for Rare Disease: A Case Study to Stratify Infection Risk in Pediatric Leukemia. J Clin Med 2024; 13:1788. [PMID: 38542012 PMCID: PMC10970787 DOI: 10.3390/jcm13061788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 04/18/2024] Open
Abstract
Background: Datasets on rare diseases, like pediatric acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL), have small sample sizes that hinder machine learning (ML). The objective was to develop an interpretable ML framework to elucidate actionable insights from small tabular rare disease datasets. Methods: The comprehensive framework employed optimized data imputation and sampling, supervised and unsupervised learning, and literature-based discovery (LBD). The framework was deployed to assess treatment-related infection in pediatric AML and ALL. Results: An interpretable decision tree classified the risk of infection as either "high risk" or "low risk" in pediatric ALL (n = 580) and AML (n = 132) with accuracy of ∼79%. Interpretable regression models predicted the discrete number of developed infections with a mean absolute error (MAE) of 2.26 for bacterial infections and an MAE of 1.29 for viral infections. Features that best explained the development of infection were the chemotherapy regimen, cancer cells in the central nervous system at initial diagnosis, chemotherapy course, leukemia type, Down syndrome, race, and National Cancer Institute risk classification. Finally, SemNet 2.0, an open-source LBD software that links relationships from 33+ million PubMed articles, identified additional features for the prediction of infection, like glucose, iron, neutropenia-reducing growth factors, and systemic lupus erythematosus (SLE). Conclusions: The developed ML framework enabled state-of-the-art, interpretable predictions using rare disease tabular datasets. ML model performance baselines were successfully produced to predict infection in pediatric AML and ALL.
Collapse
Affiliation(s)
- Irfan Al-Hussaini
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Brandon White
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Armon Varmeziar
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Nidhi Mehra
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Milagro Sanchez
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Judy Lee
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA (T.P.M.)
| | - Nicholas P. DeGroote
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA (T.P.M.)
| | - Tamara P. Miller
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA (T.P.M.)
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Emory University, Atlanta, GA 30332, USA
| | - Cassie S. Mitchell
- Laboratory for Pathology Dynamics, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Machine Learning Center at Georgia Tech, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
30
|
Xu X, Qi Z, Wang L, Zhang M, Geng Z, Han X. Gsw-fi: a GLM model incorporating shrinkage and double-weighted strategies for identifying cancer driver genes with functional impact. BMC Bioinformatics 2024; 25:99. [PMID: 38448819 PMCID: PMC10916024 DOI: 10.1186/s12859-024-05707-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Cancer, a disease with high morbidity and mortality rates, poses a significant threat to human health. Driver genes, which harbor mutations accountable for the initiation and progression of tumors, play a crucial role in cancer development. Identifying driver genes stands as a paramount objective in cancer research and precision medicine. RESULTS In the present work, we propose a method for identifying driver genes using a Generalized Linear Regression Model (GLM) with Shrinkage and double-Weighted strategies based on Functional Impact, which is named GSW-FI. Firstly, an estimating model is proposed for assessing the background functional impacts of genes based on GLM, utilizing gene features as predictors. Secondly, the shrinkage and double-weighted strategies as two revising approaches are integrated to ensure the rationality of the identified driver genes. Lastly, a statistical method of hypothesis testing is designed to identify driver genes by leveraging the estimated background function impacts. Experimental results conducted on 31 The Cancer Genome Altas datasets demonstrate that GSW-FI outperforms ten other prediction methods in terms of the overlap fraction with well-known databases and consensus predictions among different methods. CONCLUSIONS GSW-FI presents a novel approach that efficiently identifies driver genes with functional impact mutations using computational methods, thereby advancing the development of precision medicine for cancer.
Collapse
Affiliation(s)
- Xiaolu Xu
- School of Computer and Artificial Intelligence, Liaoning Normal University, Dalian, China
| | - Zitong Qi
- Department of Statistics, University of Washington, Seattle, USA
| | - Lei Wang
- Center for Reproductive and Genetic Medicine, Dalian Women and Children's Medical Group, Dalian, China.
| | - Meiwei Zhang
- Center for Reproductive and Genetic Medicine, Dalian Women and Children's Medical Group, Dalian, China.
| | - Zhaohong Geng
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiumei Han
- College of Artificial Intelligence, Dalian Maritime University, Dalian, China
| |
Collapse
|
31
|
Shu L, Yan H, Wu Y, Yan T, Yang L, Zhang S, Chen Z, Liao Q, Yang L, Xiao B, Ye M, Lv S, Wu M, Zhu X, Hu P. Explainable machine learning in outcome prediction of high-grade aneurysmal subarachnoid hemorrhage. Aging (Albany NY) 2024; 16:4654-4669. [PMID: 38431285 PMCID: PMC10968679 DOI: 10.18632/aging.205621] [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: 09/05/2023] [Accepted: 01/29/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE Accurate prognostic prediction in patients with high-grade aneruysmal subarachnoid hemorrhage (aSAH) is essential for personalized treatment. In this study, we developed an interpretable prognostic machine learning model for high-grade aSAH patients using SHapley Additive exPlanations (SHAP). METHODS A prospective registry cohort of high-grade aSAH patients was collected in one single-center hospital. The endpoint in our study is a 12-month follow-up outcome. The dataset was divided into training and validation sets in a 7:3 ratio. Machine learning algorithms, including Logistic regression model (LR), support vector machine (SVM), random forest (RF), and extreme gradient boosting (XGBoost), were employed to develop a prognostic prediction model for high-grade aSAH. The optimal model was selected for SHAP analysis. RESULTS Among the 421 patients, 204 (48.5%) exhibited poor prognosis. The RF model demonstrated superior performance compared to LR (AUC = 0.850, 95% CI: 0.783-0.918), SVM (AUC = 0.862, 95% CI: 0.799-0.926), and XGBoost (AUC = 0.850, 95% CI: 0.783-0.917) with an AUC of 0.867 (95% CI: 0.806-0 .929). Primary prognostic features identified through SHAP analysis included higher World Federation of Neurosurgical Societies (WFNS) grade, higher modified Fisher score (mFS) and advanced age, were found to be associated with 12-month unfavorable outcome, while the treatment of coiling embolization for aSAH drove the prediction towards favorable prognosis. Additionally, the SHAP force plot visualized individual prognosis predictions. CONCLUSIONS This study demonstrated the potential of machine learning techniques in prognostic prediction for high-grade aSAH patients. The features identified through SHAP analysis enhance model interpretability and provide guidance for clinical decision-making.
Collapse
Affiliation(s)
- Lei Shu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Hua Yan
- Department of Emergency, Affiliated Hospital of Panzhihua University, Panzhihua 617000, Sichuan, China
| | - Yanze Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Tengfeng Yan
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Li Yang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Si Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Zhihao Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Qiuye Liao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Lu Yang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Bing Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Minhua Ye
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Shigang Lv
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Miaojing Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Ping Hu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, China
- Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| |
Collapse
|
32
|
Bhatia K, Sandhu V, Wong MH, Iyer P, Bhatt S. Therapeutic biomarkers in acute myeloid leukemia: functional and genomic approaches. Front Oncol 2024; 14:1275251. [PMID: 38410111 PMCID: PMC10894932 DOI: 10.3389/fonc.2024.1275251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/17/2024] [Indexed: 02/28/2024] Open
Abstract
Acute myeloid leukemia (AML) is clinically and genetically a heterogeneous disease characterized by clonal expansion of abnormal hematopoietic progenitors. Genomic approaches to precision medicine have been implemented to direct targeted therapy for subgroups of AML patients, for instance, IDH inhibitors for IDH1/2 mutated patients, and FLT3 inhibitors with FLT3 mutated patients. While next generation sequencing for genetic mutations has improved treatment outcomes, only a fraction of AML patients benefit due to the low prevalence of actionable targets. In recent years, the adoption of newer functional technologies for quantitative phenotypic analysis and patient-derived avatar models has strengthened the potential for generalized functional precision medicine approach. However, functional approach requires robust standardization for multiple variables such as functional parameters, time of drug exposure and drug concentration for making in vitro predictions. In this review, we first summarize genomic and functional therapeutic biomarkers adopted for AML therapy, followed by challenges associated with these approaches, and finally, the future strategies to enhance the implementation of precision medicine.
Collapse
Affiliation(s)
- Karanpreet Bhatia
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Vedant Sandhu
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Mei Hsuan Wong
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Prasad Iyer
- Children's Blood and Cancer Centre, KK Women's and Children's Hospital, Singapore, Singapore
- Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Shruti Bhatt
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| |
Collapse
|
33
|
Qian Y, Shi M, Zhang Q. CONSMI: Contrastive Learning in the Simplified Molecular Input Line Entry System Helps Generate Better Molecules. Molecules 2024; 29:495. [PMID: 38276573 PMCID: PMC10821140 DOI: 10.3390/molecules29020495] [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: 11/15/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
In recent years, the application of deep learning in molecular de novo design has gained significant attention. One successful approach involves using SMILES representations of molecules and treating the generation task as a text generation problem, yielding promising results. However, the generation of more effective and novel molecules remains a key research area. Due to the fact that a molecule can have multiple SMILES representations, it is not sufficient to consider only one of them for molecular generation. To make up for this deficiency, and also motivated by the advancements in contrastive learning in natural language processing, we propose a contrastive learning framework called CONSMI to learn more comprehensive SMILES representations. This framework leverages different SMILES representations of the same molecule as positive examples and other SMILES representations as negative examples for contrastive learning. The experimental results of generation tasks demonstrate that CONSMI significantly enhances the novelty of generated molecules while maintaining a high validity. Moreover, the generated molecules have similar chemical properties compared to the original dataset. Additionally, we find that CONSMI can achieve favorable results in classifier tasks, such as the compound-protein interaction task.
Collapse
Affiliation(s)
| | | | - Qian Zhang
- School of Computer Science and Technology, Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, East China Normal University, 3663 North Zhongshan Road, Putuo District, Shanghai 200062, China; (Y.Q.); (M.S.)
| |
Collapse
|
34
|
DeGroat W, Abdelhalim H, Patel K, Mendhe D, Zeeshan S, Ahmed Z. Discovering biomarkers associated and predicting cardiovascular disease with high accuracy using a novel nexus of machine learning techniques for precision medicine. Sci Rep 2024; 14:1. [PMID: 38167627 PMCID: PMC10762256 DOI: 10.1038/s41598-023-50600-8] [Citation(s) in RCA: 131] [Impact Index Per Article: 131.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
Personalized interventions are deemed vital given the intricate characteristics, advancement, inherent genetic composition, and diversity of cardiovascular diseases (CVDs). The appropriate utilization of artificial intelligence (AI) and machine learning (ML) methodologies can yield novel understandings of CVDs, enabling improved personalized treatments through predictive analysis and deep phenotyping. In this study, we proposed and employed a novel approach combining traditional statistics and a nexus of cutting-edge AI/ML techniques to identify significant biomarkers for our predictive engine by analyzing the complete transcriptome of CVD patients. After robust gene expression data pre-processing, we utilized three statistical tests (Pearson correlation, Chi-square test, and ANOVA) to assess the differences in transcriptomic expression and clinical characteristics between healthy individuals and CVD patients. Next, the recursive feature elimination classifier assigned rankings to transcriptomic features based on their relation to the case-control variable. The top ten percent of commonly observed significant biomarkers were evaluated using four unique ML classifiers (Random Forest, Support Vector Machine, Xtreme Gradient Boosting Decision Trees, and k-Nearest Neighbors). After optimizing hyperparameters, the ensembled models, which were implemented using a soft voting classifier, accurately differentiated between patients and healthy individuals. We have uncovered 18 transcriptomic biomarkers that are highly significant in the CVD population that were used to predict disease with up to 96% accuracy. Additionally, we cross-validated our results with clinical records collected from patients in our cohort. The identified biomarkers served as potential indicators for early detection of CVDs. With its successful implementation, our newly developed predictive engine provides a valuable framework for identifying patients with CVDs based on their biomarker profiles.
Collapse
Affiliation(s)
- William DeGroat
- Health Care Policy and Aging Research, Rutgers Institute for Health, Rutgers University, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Habiba Abdelhalim
- Health Care Policy and Aging Research, Rutgers Institute for Health, Rutgers University, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Kush Patel
- Health Care Policy and Aging Research, Rutgers Institute for Health, Rutgers University, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Dinesh Mendhe
- Health Care Policy and Aging Research, Rutgers Institute for Health, Rutgers University, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Saman Zeeshan
- Rutgers Cancer Institute of New Jersey, Rutgers University, 195 Little Albany St, New Brunswick, NJ, USA
| | - Zeeshan Ahmed
- Health Care Policy and Aging Research, Rutgers Institute for Health, Rutgers University, 112 Paterson St, New Brunswick, NJ, 08901, USA.
- Department of Medicine/Cardiovascular Disease and Hypertension, Robert Wood Johnson Medical School, Rutgers Biomedical and Health Sciences, 125 Paterson St, New Brunswick, NJ, USA.
| |
Collapse
|
35
|
Szeremeta M, Janica J, Niemcunowicz-Janica A. Artificial intelligence in forensic medicine and related sciences - selected issues. ARCHIVES OF FORENSIC MEDICINE AND CRIMINOLOGY 2024; 74:64-76. [PMID: 39450596 DOI: 10.4467/16891716amsik.24.005.19650] [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: 02/23/2024] [Accepted: 04/16/2024] [Indexed: 10/26/2024] Open
Abstract
Aim The aim of the work is to provide an overview of the potential application of artificial intelligence in forensic medicine and related sciences, and to identify concerns related to providing medico-legal opinions and legal liability in cases in which possible harm in terms of diagnosis and/or treatment is likely to occur when using an advanced system of computer-based information processing and analysis. Material and methods The material for the study comprised scientific literature related to the issue of artificial intelligence in forensic medicine and related sciences. For this purpose, Google Scholar, PubMed and ScienceDirect databases were searched. To identify useful articles, such terms as "artificial intelligence," "deep learning," "machine learning," "forensic medicine," "legal medicine," "forensic pathology" and "medicine" were used. In some cases, articles were identified based on the semantic proximity of the introduced terms. Conclusions Dynamic development of the computing power and the ability of artificial intelligence to analyze vast data volumes made it possible to transfer artificial intelligence methods to forensic medicine and related sciences. Artificial intelligence has numerous applications in forensic medicine and related sciences and can be helpful in thanatology, forensic traumatology, post-mortem identification examinations, as well as post-mortem microscopic and toxicological diagnostics. Analyzing the legal and medico-legal aspects, artificial intelligence in medicine should be treated as an auxiliary tool, whereas the final diagnostic and therapeutic decisions and the extent to which they are implemented should be the responsibility of humans.
Collapse
Affiliation(s)
- Michał Szeremeta
- Department of Forensic Medicine, Medical University of Białystok, Poland
| | - Julia Janica
- Student's Scientific Group at the Department of Forensic Medicine, Poland
| | | |
Collapse
|
36
|
Zuhair V, Babar A, Ali R, Oduoye MO, Noor Z, Chris K, Okon II, Rehman LU. Exploring the Impact of Artificial Intelligence on Global Health and Enhancing Healthcare in Developing Nations. J Prim Care Community Health 2024; 15:21501319241245847. [PMID: 38605668 PMCID: PMC11010755 DOI: 10.1177/21501319241245847] [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: 10/19/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Artificial intelligence (AI), which combines computer science with extensive datasets, seeks to mimic human-like intelligence. Subsets of AI are being applied in almost all fields of medicine and surgery. AIM This review focuses on the applications of AI in healthcare settings in developing countries, designed to underscore its significance by comprehensively outlining the advancements made thus far, the shortcomings encountered in AI applications, the present status of AI integration, persistent challenges, and innovative strategies to surmount them. METHODOLOGY Articles from PubMed, Google Scholar, and Cochrane were searched from 2000 to 2023 with keywords including AI and healthcare, focusing on multiple medical specialties. RESULTS The increasing role of AI in diagnosis, prognosis prediction, and patient management, as well as hospital management and community healthcare, has made the overall healthcare system more efficient, especially in the high patient load setups and resource-limited areas of developing countries where patient care is often compromised. However, challenges, including low adoption rates and the absence of standardized guidelines, high installation and maintenance costs of equipment, poor transportation and connectivvity issues hinder AI's full use in healthcare. CONCLUSION Despite these challenges, AI holds a promising future in healthcare. Adequate knowledge and expertise of healthcare professionals for the use of AI technology in healthcare is imperative in developing nations.
Collapse
Affiliation(s)
- Varisha Zuhair
- Jinnah Sindh Medical University, Karachi, Sindh, Pakistan
| | - Areesha Babar
- Jinnah Sindh Medical University, Karachi, Sindh, Pakistan
| | - Rabbiya Ali
- Jinnah Sindh Medical University, Karachi, Sindh, Pakistan
| | - Malik Olatunde Oduoye
- The Medical Research Circle, (MedReC), Gisenyi, Goma, Democratic Republic of the Congo
| | - Zainab Noor
- Institute of Dentistry CMH Lahore Medical College, Lahore, Punjab, Pakistan
| | - Kitumaini Chris
- The Medical Research Circle, (MedReC), Gisenyi, Goma, Democratic Republic of the Congo
- Université Libre des Pays des Grands-Lacs Goma, Noth-Kivu, Democratic Republic of the Congo
| | - Inibehe Ime Okon
- The Medical Research Circle, (MedReC), Gisenyi, Goma, Democratic Republic of the Congo
- NiMSA SCOPH, Uyo, Akwa-Ibom State, Nigeria
| | | |
Collapse
|
37
|
Pagallo U, O’Sullivan S, Nevejans N, Holzinger A, Friebe M, Jeanquartier F, Jean-Quartier C, Miernik A. The underuse of AI in the health sector: Opportunity costs, success stories, risks and recommendations. HEALTH AND TECHNOLOGY 2023; 14:1-14. [PMID: 38229886 PMCID: PMC10788319 DOI: 10.1007/s12553-023-00806-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/16/2023] [Indexed: 01/18/2024]
Abstract
Purpose This contribution explores the underuse of artificial intelligence (AI) in the health sector, what this means for practice, and how much the underuse can cost. Attention is drawn to the relevance of an issue that the European Parliament has outlined as a "major threat" in 2020. At its heart is the risk that research and development on trusted AI systems for medicine and digital health will pile up in lab centers without generating further practical relevance. Our analysis highlights why researchers, practitioners and especially policymakers, should pay attention to this phenomenon. Methods The paper examines the ways in which governments and public agencies are addressing the underuse of AI. As governments and international organizations often acknowledge the limitations of their own initiatives, the contribution explores the causes of the current issues and suggests ways to improve initiatives for digital health. Results Recommendations address the development of standards, models of regulatory governance, assessment of the opportunity costs of underuse of technology, and the urgency of the problem. Conclusions The exponential pace of AI advances and innovations makes the risks of underuse of AI increasingly threatening. Graphical Abstract
Collapse
Affiliation(s)
- Ugo Pagallo
- Law School, University of Turin, Turin, Italy
| | - Shane O’Sullivan
- Department of Urology, Faculty of Medicine, University of Freiburg - Medical Centre, Freiburg im Breisgau, Germany
| | - Nathalie Nevejans
- Ethics and Procedures Center (CDEP), Faculty of Law of Douai, University of Artois, Arras, France
| | - Andreas Holzinger
- Human-Centered AI Lab, Medical University of Graz, Graz, Austria
- University of Natural Resources and Life Sciences Vienna, Human-Centered AI Lab, Vienna, Austria
| | - Michael Friebe
- Department of Measurements and Electronics, AGH University of Science and Technology, Krak’ow, Poland
- Faculty of Medicine, Otto-von-Guericke-University, Magdeburg, Germany
- Center for Innovation and Business Development, FOM University of Applied Sciences, Essen, Germany
| | | | | | - Arkadiusz Miernik
- Department of Urology, Faculty of Medicine, University of Freiburg - Medical Centre, Freiburg im Breisgau, Germany
| |
Collapse
|
38
|
Hu ZT, Yu Y, Chen R, Yeh SJ, Chen B, Huang H. Large-Scale Information Retrieval and Correction of Noisy Pharmacogenomic Datasets through Residual Thresholded Deep Matrix Factorization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.07.570723. [PMID: 38106027 PMCID: PMC10723412 DOI: 10.1101/2023.12.07.570723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Pharmacogenomics studies are attracting an increasing amount of interest from researchers in precision medicine. The advances in high-throughput experiments and multiplexed approaches allow the large-scale quantification of drug sensitivities in molecularly characterized cancer cell lines (CCLs), resulting in a number of open drug sensitivity datasets for drug biomarker discovery. However, a significant inconsistency in drug sensitivity values among these datasets has been noted. Such inconsistency indicates the presence of substantial noise, subsequently hindering downstream analyses. To address the noise in drug sensitivity data, we introduce a robust and scalable deep learning framework, Residual Thresholded Deep Matrix Factorization (RT-DMF). This method takes a single drug sensitivity data matrix as its sole input and outputs a corrected and imputed matrix. Deep Matrix Factorization (DMF) excels at uncovering subtle patterns, due to its minimal reliance on data structure assumptions. This attribute significantly boosts DMF's ability to identify complex hidden patterns among nuisance effects in the data, thereby facilitating the detection of signals that are therapeutically relevant. Furthermore, RT-DMF incorporates an iterative residual thresholding (RT) procedure, which plays a crucial role in retaining signals more likely to hold therapeutic importance. Validation using simulated datasets and real pharmacogenomics datasets demonstrates the effectiveness of our approach in correcting noise and imputing missing data in drug sensitivity datasets (open source package available at https://github.com/tomwhoooo/rtdmf).
Collapse
Affiliation(s)
- Zhiyue Tom Hu
- Division of Biostatistics, University of California Berkeley, Berkeley, 94720, U.S.A
| | - Yaodong Yu
- Department of Electrical Engineer and Computer Science, University of California Berkeley, Berkeley, 94720, U.S.A
| | - Ruoqiao Chen
- Department of Pharmacology and Toxicology, Michigan State University, 48824, U.S.A
| | - Shan-Ju Yeh
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan R.O.C
| | - Bin Chen
- Department of Pharmacology and Toxicology, Michigan State University, 48824, U.S.A
- Department of Pediatrics and Human Development, Michigan State University, 48824, U.S.A
| | - Haiyan Huang
- Department of Statistics, University of California Berkeley, Berkeley, 94720, U.S.A
| |
Collapse
|
39
|
Carmina D, Benfenati V, Simonelli C, Rotolo A, Cardano P, Grovale N, Mangoni di S Stefano L, de Santo T, Zamboni R, Palermo V, Muccini M, De Seta F. Innovative solutions for disease management. Bioelectron Med 2023; 9:28. [PMID: 38053220 DOI: 10.1186/s42234-023-00131-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
The increasing prevalence of chronic diseases is a driver for emerging big data technologies for healthcare including digital platforms for data collection, systems for active patient engagement and education, therapy specific predictive models, optimized patient pathway models. Powerful bioelectronic medicine tools for data collection, analysis and visualization allow for joint processing of large volumes of heterogeneous data, which in turn can produce new insights about patient outcomes and alternative interpretations of clinical patterns that can lead to implementation of optimized clinical decisions and clinical patient pathway by healthcare professionals.With this perspective, we identify innovative solutions for disease management and evaluate their impact on patients, payers and society, by analyzing their impact in terms of clinical outcomes (effectiveness, safety, and quality of life) and economic outcomes (cost-effectiveness, savings, and productivity).As a result, we propose a new approach based on the main pillars of innovation in the disease management area, i.e. progressive patient care models, patient-centric approaches, bioelectronics for precise medicine, and lean management that, combined with an increase in appropriate private-public-citizen-partnership, leads towards Patient-Centric Healthcare.
Collapse
Affiliation(s)
- Dafni Carmina
- Medtronic Clinical & Regulatory Solutions - Study & Scientific Solutions, Via Aurelia 866, Roma, 00165, Italy.
| | - Valentina Benfenati
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e Fotoreattività, via Gobetti 101, Bologna, 40129, Italy.
| | - Claudia Simonelli
- Medtronic Clinical & Regulatory Solutions - Study & Scientific Solutions, Via Aurelia 866, Roma, 00165, Italy
| | - Alessia Rotolo
- Consiglio Nazionale delle Ricerche, Istituto per lo Studio dei Materiali Nanostrutturati, via Gobetti 101, Bologna, 40129, Italy
| | - Paola Cardano
- Medtronic Clinical & Regulatory Solutions - Study & Scientific Solutions, Via Aurelia 866, Roma, 00165, Italy
| | - Nicoletta Grovale
- Medtronic Clinical & Regulatory Solutions - Study & Scientific Solutions, Via Aurelia 866, Roma, 00165, Italy
| | | | - Tiziana de Santo
- Medtronic Clinical & Regulatory Solutions - Study & Scientific Solutions, Via Aurelia 866, Roma, 00165, Italy
| | - Roberto Zamboni
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e Fotoreattività, via Gobetti 101, Bologna, 40129, Italy
| | - Vincenzo Palermo
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e Fotoreattività, via Gobetti 101, Bologna, 40129, Italy
| | - Michele Muccini
- Consiglio Nazionale delle Ricerche, Istituto per lo Studio dei Materiali Nanostrutturati, via Gobetti 101, Bologna, 40129, Italy
- Mister Smart Innovation S, via Gobetti 101, Bologna, 40129, Italy
| | - Francesco De Seta
- Medtronic Clinical & Regulatory Solutions - Study & Scientific Solutions, Via Aurelia 866, Roma, 00165, Italy
| |
Collapse
|
40
|
Aribi A, Salhotra A, Afkhami M, Munteanu A, Ali H, Aldoss I, Otoukesh S, Al Malki MM, Sandhu KS, Koller P, Arslan S, Stewart F, Artz A, Curtin P, Ball B, O'Hearn J, Spielberger R, Smith E, Budde E, Nakamura R, Stein A, Forman S, Marcucci G, Becker PS, Pullarkat V. WT1-mutated acute myeloid leukemia is sensitive to fludarabine-based chemotherapy and conditioning regimens. Leuk Lymphoma 2023; 64:1811-1821. [PMID: 37533373 DOI: 10.1080/10428194.2023.2241096] [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: 03/10/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023]
Abstract
We conducted a retrospective analysis of WT1-mutated acute myeloid leukemia (AML) patients who underwent allogeneic stem cell transplant. Thirty-seven patients with WT1-mutated AML were identified. Primary induction failure (40%) and early relapse rate (18%) after idarubicin/cytarabine (7 + 3) chemotherapy were observed. All patients with induction failure subsequently achieved CR with additional chemotherapy. There was no significant difference between outcomes after myeloablative vs. reduced intensity (Fludarabine/Melphalan [Flu/Mel]) conditioning regimens. RFS but not OS was significantly better in patients who received FLAG-IDA prior to transplant and/or a fludarabine-containing conditioning. In an independent ex vivo study, WT1-mutated AML samples exhibited greater sensitivity to fludarabine (p = 0.026) and melphalan (p = 0.0005) than non-WT1-mutated AML samples while there was no difference between sensitivity to cytarabine. Our data favor using a fludarabine-based induction for AML with WT1 mutation instead of 7 + 3. Fludarabine conditioning regimens for alloHCT showed better RFS but not OS.
Collapse
Affiliation(s)
- Ahmed Aribi
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Amandeep Salhotra
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Michelle Afkhami
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Anamaria Munteanu
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Haris Ali
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Ibrahim Aldoss
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Salman Otoukesh
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Monzr M Al Malki
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Karamjeet S Sandhu
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Paul Koller
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Shukaib Arslan
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Forrest Stewart
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Andrew Artz
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Peter Curtin
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Brian Ball
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - James O'Hearn
- Department of Clinical Translational Project Development, City of Hope National Medical Center, Duarte, CA, USA
| | - Ricardo Spielberger
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Eileen Smith
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Elizabeth Budde
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Ryotaro Nakamura
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Anthony Stein
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Stephen Forman
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Guido Marcucci
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Pamela S Becker
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| | - Vinod Pullarkat
- Department of Hematology, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
41
|
Garg P, Mohanty A, Ramisetty S, Kulkarni P, Horne D, Pisick E, Salgia R, Singhal SS. Artificial intelligence and allied subsets in early detection and preclusion of gynecological cancers. Biochim Biophys Acta Rev Cancer 2023; 1878:189026. [PMID: 37980945 DOI: 10.1016/j.bbcan.2023.189026] [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: 09/17/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 11/21/2023]
Abstract
Gynecological cancers including breast, cervical, ovarian, uterine, and vaginal, pose the greatest threat to world health, with early identification being crucial to patient outcomes and survival rates. The application of machine learning (ML) and artificial intelligence (AI) approaches to the study of gynecological cancer has shown potential to revolutionize cancer detection and diagnosis. The current review outlines the significant advancements, obstacles, and prospects brought about by AI and ML technologies in the timely identification and accurate diagnosis of different types of gynecological cancers. The AI-powered technologies can use genomic data to discover genetic alterations and biomarkers linked to a particular form of gynecologic cancer, assisting in the creation of targeted treatments. Furthermore, it has been shown that the potential benefits of AI and ML technologies in gynecologic tumors can greatly increase the accuracy and efficacy of cancer diagnosis, reduce diagnostic delays, and possibly eliminate the need for needless invasive operations. In conclusion, the review focused on the integrative part of AI and ML based tools and techniques in the early detection and exclusion of various cancer types; together with a collaborative coordination between research clinicians, data scientists, and regulatory authorities, which is suggested to realize the full potential of AI and ML in gynecologic cancer care.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Atish Mohanty
- Departments of Medical Oncology & Therapeutics Research, Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sravani Ramisetty
- Departments of Medical Oncology & Therapeutics Research, Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Departments of Medical Oncology & Therapeutics Research, Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Evan Pisick
- Department of Medical Oncology, City of Hope, Chicago, IL 60099, USA
| | - Ravi Salgia
- Departments of Medical Oncology & Therapeutics Research, Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S Singhal
- Departments of Medical Oncology & Therapeutics Research, Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
42
|
Xu X, Ma W, Qiu G, Xuan L, He C, Zhang T, Wang J, Liu Q. Venetoclax Overcomes Sorafenib Resistance in Acute Myeloid Leukemia by Targeting BCL2. BIOLOGY 2023; 12:1337. [PMID: 37887047 PMCID: PMC10603903 DOI: 10.3390/biology12101337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023]
Abstract
Sorafenib, a kinase inhibitor, has shown promising therapeutic efficacy in a subset of patients with acute myeloid leukemia (AML). However, despite its clinical effectiveness, sorafenib resistance is frequently observed in clinical settings, and the mechanisms underlying this resistance as well as effective strategies to overcome it remain unclear. We examined both single-cell and bulk transcription data in sorafenib-resistant and control AML patients and integrated a sorafenib resistance gene signature to predict the sensitivity of AML cells and the clinical outcomes of AML patients undergoing sorafenib therapy. In addition, our drug sensitivity analysis of scRNA-seq data using deconvolution methods showed that venetoclax was effective in targeting sorafenib-resistant AML cells. Mechanistically, sorafenib was found to activate the JAK-STAT3 pathway and upregulate BCL2 expression in sorafenib-resistant AML cells. This upregulation of BCL2 expression rendered the cells vulnerable to the BCL2 inhibitor venetoclax. In conclusion, we developed a platform to predict sorafenib resistance and clinical outcomes in AML patients after therapy. Our findings suggest that the combination of sorafenib and venetoclax could be an effective therapeutic strategy for AML treatment.
Collapse
Affiliation(s)
- Xi Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510091, China (L.X.)
| | - Weiwei Ma
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China;
| | - Guo Qiu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510091, China (L.X.)
| | - Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510091, China (L.X.)
| | - Chong He
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou 510080, China
| | - Tian Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510091, China (L.X.)
| | - Jian Wang
- Children’s Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510091, China (L.X.)
| |
Collapse
|
43
|
Dragan P, Joshi K, Atzei A, Latek D. Keras/TensorFlow in Drug Design for Immunity Disorders. Int J Mol Sci 2023; 24:15009. [PMID: 37834457 PMCID: PMC10573944 DOI: 10.3390/ijms241915009] [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: 09/08/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Homeostasis of the host immune system is regulated by white blood cells with a variety of cell surface receptors for cytokines. Chemotactic cytokines (chemokines) activate their receptors to evoke the chemotaxis of immune cells in homeostatic migrations or inflammatory conditions towards inflamed tissue or pathogens. Dysregulation of the immune system leading to disorders such as allergies, autoimmune diseases, or cancer requires efficient, fast-acting drugs to minimize the long-term effects of chronic inflammation. Here, we performed structure-based virtual screening (SBVS) assisted by the Keras/TensorFlow neural network (NN) to find novel compound scaffolds acting on three chemokine receptors: CCR2, CCR3, and one CXC receptor, CXCR3. Keras/TensorFlow NN was used here not as a typically used binary classifier but as an efficient multi-class classifier that can discard not only inactive compounds but also low- or medium-activity compounds. Several compounds proposed by SBVS and NN were tested in 100 ns all-atom molecular dynamics simulations to confirm their binding affinity. To improve the basic binding affinity of the compounds, new chemical modifications were proposed. The modified compounds were compared with known antagonists of these three chemokine receptors. Known CXCR3 compounds were among the top predicted compounds; thus, the benefits of using Keras/TensorFlow in drug discovery have been shown in addition to structure-based approaches. Furthermore, we showed that Keras/TensorFlow NN can accurately predict the receptor subtype selectivity of compounds, for which SBVS often fails. We cross-tested chemokine receptor datasets retrieved from ChEMBL and curated datasets for cannabinoid receptors. The NN model trained on the cannabinoid receptor datasets retrieved from ChEMBL was the most accurate in the receptor subtype selectivity prediction. Among NN models trained on the chemokine receptor datasets, the CXCR3 model showed the highest accuracy in differentiating the receptor subtype for a given compound dataset.
Collapse
Affiliation(s)
- Paulina Dragan
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-903 Warsaw, Poland; (P.D.); (A.A.)
| | - Kavita Joshi
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-903 Warsaw, Poland; (P.D.); (A.A.)
| | - Alessandro Atzei
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-903 Warsaw, Poland; (P.D.); (A.A.)
- Department of Life and Environmental Science, Food Toxicology Unit, University of Cagliari, University Campus of Monserrato, SS 554, 09042 Cagliari, Italy
| | - Dorota Latek
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-903 Warsaw, Poland; (P.D.); (A.A.)
| |
Collapse
|
44
|
Abdallah S, Sharifa M, I Kh Almadhoun MK, Khawar MM, Shaikh U, Balabel KM, Saleh I, Manzoor A, Mandal AK, Ekomwereren O, Khine WM, Oyelaja OT. The Impact of Artificial Intelligence on Optimizing Diagnosis and Treatment Plans for Rare Genetic Disorders. Cureus 2023; 15:e46860. [PMID: 37954711 PMCID: PMC10636514 DOI: 10.7759/cureus.46860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/14/2023] Open
Abstract
Rare genetic disorders (RDs), characterized by their low prevalence and diagnostic complexities, present significant challenges to healthcare systems. This article explores the transformative impact of artificial intelligence (AI) and machine learning (ML) in addressing these challenges. It emphasizes the need for accurate and early diagnosis of RDs, often hindered by genetic and clinical heterogeneity. This article discusses how AI and ML are reshaping healthcare, providing examples of their effectiveness in disease diagnosis, prognosis, image analysis, and drug repurposing. It highlights AI's ability to efficiently analyze extensive datasets and expedite diagnosis, showcasing case studies like Face2Gene. Furthermore, the article explores how AI tailors treatment plans for RDs, leveraging ML and deep learning (DL) to create personalized therapeutic regimens. It emphasizes AI's role in drug discovery, including the identification of potential candidates for rare disease treatments. Challenges and limitations related to AI in healthcare, including ethical, legal, technical, and human aspects, are addressed. This article underscores the importance of data ethics, privacy, and algorithmic fairness, as well as the need for standardized evaluation techniques and transparency in AI research. It highlights second-generation AI systems that prioritize patient-centric care, efficient patient recruitment for clinical trials, and the significance of high-quality data. The integration of AI with telemedicine, the growth of health databases, and the potential for personalized therapeutic recommendations are identified as promising directions for the field. In summary, this article provides a comprehensive exploration of how AI and ML are revolutionizing the diagnosis and treatment of RDs, addressing challenges while considering ethical implications in this rapidly evolving healthcare landscape.
Collapse
Affiliation(s)
- Shenouda Abdallah
- Surgery, Jaber Al Ahmad Al Jaber Al Sabah Hospital, Kuwait City, KWT
| | | | | | | | - Unzla Shaikh
- Internal Medicine, Liaquat University of Medical and Health Sciences, Hyderabad, PAK
| | | | - Inam Saleh
- Pediatrics, University of Kentucky College of Medicine, Lexington, USA
| | - Amima Manzoor
- Internal Medicine, Jinnah Sindh Medical University, Karachi, PAK
| | - Arun Kumar Mandal
- General Medicine, Mahawai Basic Hospital/The Oda Foundation, Kalikot, NPL
- Medicine, Manipal College of Medical Sciences, Pokhara, NPL
| | - Osatohanmwen Ekomwereren
- Trauma and Orthopaedics, Royal Shrewsbury Hospital, Shrewsbury and Telford Hospital NHS Trust, Shrewsbury, GBR
| | - Wai Mon Khine
- Internal Medicine, Caribbean Medical School, St. Georges, GRD
| | | |
Collapse
|
45
|
Alaiti RK, Vallio CS, Assunção JH, de Andrade e Silva FB, Gracitelli MEC, Neto AAF, Malavolta EA. Using Machine Learning to Predict Nonachievement of Clinically Significant Outcomes After Rotator Cuff Repair. Orthop J Sports Med 2023; 11:23259671231206180. [PMID: 37868215 PMCID: PMC10588422 DOI: 10.1177/23259671231206180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 05/30/2023] [Indexed: 10/24/2023] Open
Abstract
Background Although some evidence suggests that machine learning algorithms may outperform classical statistical methods in prognosis prediction for several orthopaedic surgeries, to our knowledge, no study has yet used machine learning to predict patient-reported outcome measures after rotator cuff repair. Purpose To determine whether machine learning algorithms using preoperative data can predict the nonachievement of the minimal clinically important difference (MCID) of disability at 2 years after rotator cuff surgical repair with a similar performance to that of other machine learning studies in the orthopaedic surgery literature. Study Design Case-control study; Level of evidence, 3. Methods We evaluated 474 patients (n = 500 shoulders) with rotator cuff tears who underwent arthroscopic rotator cuff repair between January 2013 and April 2019. The study outcome was the difference between the preoperative and 24-month postoperative American Shoulder and Elbow Surgeons (ASES) score. A cutoff score was calculated based on the established MCID of 15.2 points to separate success (higher than the cutoff) from failure (lower than the cutoff). Routinely collected imaging, clinical, and demographic data were used to train 8 machine learning algorithms (random forest classifier; light gradient boosting machine [LightGBM]; decision tree classifier; extra trees classifier; logistic regression; extreme gradient boosting [XGBoost]; k-nearest neighbors [KNN] classifier; and CatBoost classifier). We used a random sample of 70% of patients to train the algorithms, and 30% were left for performance assessment, simulating new data. The performance of the models was evaluated with the area under the receiver operating characteristic curve (AUC). Results The AUCs for all algorithms ranged from 0.58 to 0.68. The random forest classifier and LightGBM presented the highest AUC values (0.68 [95% CI, 0.48-0.79] and 0.67 [95% CI, 0.43-0.75], respectively) of the 8 machine learning algorithms. Most of the machine learning algorithms outperformed logistic regression (AUC, 0.59 [95% CI, 0.48-0.81]); nonetheless, their performance was lower than that of other machine learning studies in the orthopaedic surgery literature. Conclusion Machine learning algorithms demonstrated some ability to predict the nonachievement of the MCID on the ASES 2 years after rotator cuff repair surgery.
Collapse
Affiliation(s)
- Rafael Krasic Alaiti
- Research, Technology, and Data Science Office, Grupo Superador, São Paulo, Brazil
- Universidade de São Paulo, São Paulo, Brazil
| | - Caio Sain Vallio
- Health Innovation, Data Science, and MLOps, Semantix, São Paulo, Brazil
| | - Jorge Henrique Assunção
- Faculdade de Medicina, Hospital das Clinicas FMUSP, Universidade de São Paulo, São Paulo, Brazil
- DASA, Hospital 9 de Julho, São Paulo, São Paulo, Brazil
| | | | | | | | - Eduardo Angeli Malavolta
- Faculdade de Medicina, Hospital das Clinicas FMUSP, Universidade de São Paulo, São Paulo, Brazil
- Hospital do Coração, São Paulo, Brazil
| |
Collapse
|
46
|
Piraianu AI, Fulga A, Musat CL, Ciobotaru OR, Poalelungi DG, Stamate E, Ciobotaru O, Fulga I. Enhancing the Evidence with Algorithms: How Artificial Intelligence Is Transforming Forensic Medicine. Diagnostics (Basel) 2023; 13:2992. [PMID: 37761359 PMCID: PMC10529115 DOI: 10.3390/diagnostics13182992] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The integration of artificial intelligence (AI) into various fields has ushered in a new era of multidisciplinary progress. Defined as the ability of a system to interpret external data, learn from it, and adapt to specific tasks, AI is poised to revolutionize the world. In forensic medicine and pathology, algorithms play a crucial role in data analysis, pattern recognition, anomaly identification, and decision making. This review explores the diverse applications of AI in forensic medicine, encompassing fields such as forensic identification, ballistics, traumatic injuries, postmortem interval estimation, forensic toxicology, and more. RESULTS A thorough review of 113 articles revealed a subset of 32 papers directly relevant to the research, covering a wide range of applications. These included forensic identification, ballistics and additional factors of shooting, traumatic injuries, post-mortem interval estimation, forensic toxicology, sexual assaults/rape, crime scene reconstruction, virtual autopsy, and medical act quality evaluation. The studies demonstrated the feasibility and advantages of employing AI technology in various facets of forensic medicine and pathology. CONCLUSIONS The integration of AI in forensic medicine and pathology offers promising prospects for improving accuracy and efficiency in medico-legal practices. From forensic identification to post-mortem interval estimation, AI algorithms have shown the potential to reduce human subjectivity, mitigate errors, and provide cost-effective solutions. While challenges surrounding ethical considerations, data security, and algorithmic correctness persist, continued research and technological advancements hold the key to realizing the full potential of AI in forensic applications. As the field of AI continues to evolve, it is poised to play an increasingly pivotal role in the future of forensic medicine and pathology.
Collapse
Affiliation(s)
| | - Ana Fulga
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania; (A.-I.P.); (C.L.M.); (O.-R.C.); (D.G.P.); (O.C.); (I.F.)
| | | | | | | | - Elena Stamate
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania; (A.-I.P.); (C.L.M.); (O.-R.C.); (D.G.P.); (O.C.); (I.F.)
| | | | | |
Collapse
|
47
|
Sahu VK, Ranjan A, Paul MK, Nagar S, Devarajan S, Aich J, Basu S. AI Techniques and IoT Applications Transforming the Future of Healthcare. ADVANCES IN HEALTHCARE INFORMATION SYSTEMS AND ADMINISTRATION 2023:210-233. [DOI: 10.4018/978-1-6684-5422-0.ch014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The role of artificial intelligence (AI) has advanced from an analysis and prediction tool to extending human capabilities. Currently, AI is more of a reliable assistant fueled by human experience and need of the hour in the healthcare along with simplifying daily life. AI and Internet of Things (IoT) have opened new avenues in intelligent diagnostics, drug discovery, clinical decision support, enhancing physician-patient communication, transcribing medical documents, and remote treatment. With the advent of enhanced computational power, AI has revolutionized discovery of optimal and efficient healthcare solutions and has accelerated the development of smart solutions involving IoT-based technologies. Starting from telemedicine to predict possible health disorders, AI is gaining focus to facilitate and advance healthcare solutions in developed and underdeveloped countries. This chapter deals with the scope of AI in the present scenario to future developments as AI will soon surpass human and poses threat pertaining to misuse of cognitive sciences development.
Collapse
Affiliation(s)
- Vishal Kumar Sahu
- Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - Amit Ranjan
- Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - Manash K. Paul
- Division of Pulmonary and Critical Care Medicine, University of California, Los Angeles, USA
| | - Shuchi Nagar
- Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - Shine Devarajan
- School of Biotechnology and Bioinformatics, D.Y. Patil University (Deemed), Navi Mumbai, India
| | - Jyotirmoi Aich
- School of Biotechnology and Bioinformatics, D.Y. Patil University (Deemed), Navi Mumbai, India
| | - Soumya Basu
- Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, India
| |
Collapse
|
48
|
Janizek JD, Dincer AB, Celik S, Chen H, Chen W, Naxerova K, Lee SI. Uncovering expression signatures of synergistic drug responses via ensembles of explainable machine-learning models. Nat Biomed Eng 2023; 7:811-829. [PMID: 37127711 PMCID: PMC11149694 DOI: 10.1038/s41551-023-01034-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 04/01/2023] [Indexed: 05/03/2023]
Abstract
Machine learning may aid the choice of optimal combinations of anticancer drugs by explaining the molecular basis of their synergy. By combining accurate models with interpretable insights, explainable machine learning promises to accelerate data-driven cancer pharmacology. However, owing to the highly correlated and high-dimensional nature of transcriptomic data, naively applying current explainable machine-learning strategies to large transcriptomic datasets leads to suboptimal outcomes. Here by using feature attribution methods, we show that the quality of the explanations can be increased by leveraging ensembles of explainable machine-learning models. We applied the approach to a dataset of 133 combinations of 46 anticancer drugs tested in ex vivo tumour samples from 285 patients with acute myeloid leukaemia and uncovered a haematopoietic-differentiation signature underlying drug combinations with therapeutic synergy. Ensembles of machine-learning models trained to predict drug combination synergies on the basis of gene-expression data may improve the feature attribution quality of complex machine-learning models.
Collapse
Affiliation(s)
- Joseph D Janizek
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Ayse B Dincer
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Safiye Celik
- Recursion Pharmaceuticals, Salt Lake City, UT, USA
| | - Hugh Chen
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - William Chen
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Kamila Naxerova
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Su-In Lee
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
49
|
Banerjee J, Taroni JN, Allaway RJ, Prasad DV, Guinney J, Greene C. Machine learning in rare disease. Nat Methods 2023:10.1038/s41592-023-01886-z. [PMID: 37248386 DOI: 10.1038/s41592-023-01886-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/22/2023] [Indexed: 05/31/2023]
Abstract
High-throughput profiling methods (such as genomics or imaging) have accelerated basic research and made deep molecular characterization of patient samples routine. These approaches provide a rich portrait of genes, molecular pathways and cell types involved in disease phenotypes. Machine learning (ML) can be a useful tool for extracting disease-relevant patterns from high-dimensional datasets. However, depending upon the complexity of the biological question, machine learning often requires many samples to identify recurrent and biologically meaningful patterns. Rare diseases are inherently limited in clinical cases, leading to few samples to study. In this Perspective, we outline the challenges and emerging solutions for using ML for small sample sets, specifically in rare diseases. Advances in ML methods for rare diseases are likely to be informative for applications beyond rare diseases for which few samples exist with high-dimensional data. We propose that the method community prioritize the development of ML techniques for rare disease research.
Collapse
Affiliation(s)
| | - Jaclyn N Taroni
- Childhood Cancer Data Lab, Alex's Lemonade Stand Foundation, Philadelphia, PA, USA
| | | | | | | | - Casey Greene
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
50
|
Obasa AE, Palk AC. Responsible application of artificial intelligence in health care. S AFR J SCI 2023; 119:14889. [PMID: 39328370 PMCID: PMC11426230 DOI: 10.17159/sajs.2023/14889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/05/2023] [Indexed: 09/28/2024] Open
Affiliation(s)
- Adetayo E Obasa
- Centre for Medical Ethics and Law, WHO Bioethics Collaborating Centre, Department of Medicine, Stellenbosch University, Cape Town, South Africa
| | - Andrea C Palk
- Unit for Bioethics, Centre for Applied Ethics, Philosophy Department, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|