1
|
Bajinka O, Ouedraogo SY, Li N, Zhan X. Big data for neuroscience in the context of predictive, preventive, and personalized medicine. EPMA J 2025; 16:17-35. [PMID: 39991094 PMCID: PMC11842698 DOI: 10.1007/s13167-024-00393-1] [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: 10/28/2024] [Accepted: 12/11/2024] [Indexed: 02/25/2025]
Abstract
Accurate and precise diagnosis made the medicine the hallmark of evidence-based medicine. While attaining absolute patient satisfaction may seem impossible in the aspect of disease recurrent, personalized their mecidal conditions to their responsive treatment approach may save the day. The last generation approaches in medicine require advanced technologies that will lead to evidence-based medicine. One of the trending fields in this is the use of big data in predictive, preventive, and personalized medicine (3PM). This review dwelled through the practical examples in which big data tools harness neuroscience to add more individualized apporahes to the medical conditions in a bid to confer a more personalized treatment strategies. Moreover, the known breakthroughs of big data in 3PM, big data and 3PM in neuroscience, AI and neuroscience, limitations of big data with 3PM in neuroscience, and the challenges are thoroughly discussed. Finally, the prospects of incorporating big data in 3PM are as well discussed. The review could point out that the implications of big data in 3PM are still in their infancy and will require a holistic approach. While there is a need to carefully sensitize the community, convincing them will come under interdisciplinary and, to some extent, inter-professional collaborations, capacity building for professionals, and optimal coordination of the joint systems.
Collapse
Affiliation(s)
- Ousman Bajinka
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Serge Yannick Ouedraogo
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Na Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingao Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
2
|
Suzuki H, Fujiwara N, Singal AG, Baumert TF, Chung RT, Kawaguchi T, Hoshida Y. Prevention of liver cancer in the era of next-generation antivirals and obesity epidemic. Hepatology 2025:01515467-990000000-01139. [PMID: 39808821 PMCID: PMC7617594 DOI: 10.1097/hep.0000000000001227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 01/16/2025]
Abstract
Preventive interventions are expected to substantially improve the prognosis of patients with primary liver cancer, predominantly HCC and cholangiocarcinoma. HCC prevention is challenging in the face of the evolving etiological landscape, particularly the sharp increase in obesity-associated metabolic disorders, including metabolic dysfunction-associated steatotic liver disease. Next-generation anti-HCV and HBV drugs have substantially reduced, but not eliminated, the risk of HCC and have given way to new challenges in identifying at-risk patients. The recent development of new therapeutic agents and modalities has opened unprecedented opportunities to refine primary, secondary, and tertiary HCC prevention strategies. For primary prevention (before exposure to risk factors), public health policies, such as universal HBV vaccination, have had a substantial prognostic impact. Secondary prevention (after or during active exposure to risk factors) includes regular HCC screening and chemoprevention. Emerging biomarkers and imaging modalities for HCC risk stratification and detection may enable individual risk-based personalized and cost-effective HCC screening. Clinical studies have suggested the potential utility of lipid-lowering, antidiabetic/obesity, and anti-inflammatory agents for secondary prevention, and some of them are being evaluated in prospective clinical trials. Computational and experimental studies have identified potential chemopreventive strategies directed at diverse molecular, cellular, and systemic targets for etiology-specific and/or agnostic interventions. Tertiary prevention (in conjunction with curative-intent therapies for HCC) is an area of active research with the development of new immune-based neoadjuvant/adjuvant therapies. Cholangiocarcinoma prevention may advance with recent efforts to elucidate risk factors. These advances will collectively lead to substantial improvements in liver cancer mortality rates.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Naoto Fujiwara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Amit G. Singal
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas F. Baumert
- Inserm, U1110, Institute for Translational Medicine and Liver Diseases, University of Strasbourg, F-67000, France
- IHU Strasbourg, F-67000 Strasbourg, France
- Gastroenterology and Hepatology Service, Strasbourg University Hospitals, F-67000Strasbourg, France
| | - Raymond T. Chung
- Liver Center, GI Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Yujin Hoshida
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
3
|
Drmić A, Saccà R, Vetter T, Ehmann F. Identifying and overcoming challenges in the EMA's qualification of novel methodologies: a two-year review. Front Pharmacol 2024; 15:1470908. [PMID: 39629079 PMCID: PMC11612500 DOI: 10.3389/fphar.2024.1470908] [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: 10/28/2024] [Indexed: 12/06/2024] Open
Abstract
The EMA Qualification of Novel Methodologies procedure qualifies methods, technologies and methodologies within a well-defined context of use in a pharma R&D context based on the evaluation of the presented scientific rationale and submitted data. This policy brief analyses QoNM submissions providing policy messages and recommendations to stakeholders on how to better prepare qualification applications in this regard. The recommendations include: 1. Grounding validation strategy using a current standard measure or a distribution technique. 2. Accurately represent pertinent subgroups via accurate inclusion and exclusion criteria. 3. Establish a well-defined and specific CoU with clear descriptions of the use within a development program target population and disease stage. Lastly, it emphasizes role of the QoNM procedure in advancing medicine development methodologies within the EU.
Collapse
Affiliation(s)
- Ana Drmić
- Independent Researcher, Strasbourg, France
| | - Riccardo Saccà
- Faculty of Health, Medicine and Life Sciences (FHLM), Maastricht, Netherlands
- European Medicines Agency, Amsterdam, Netherlands
| | | | - Falk Ehmann
- European Medicines Agency, Amsterdam, Netherlands
| |
Collapse
|
4
|
Ko G, Kim PG, Yoon BH, Kim J, Song W, Byeon I, Yoon J, Lee B, Kim YK. Closha 2.0: a bio-workflow design system for massive genome data analysis on high performance cluster infrastructure. BMC Bioinformatics 2024; 25:353. [PMID: 39533201 PMCID: PMC11558834 DOI: 10.1186/s12859-024-05963-8] [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: 06/24/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The explosive growth of next-generation sequencing data has resulted in ultra-large-scale datasets and significant computational challenges. As the cost of next-generation sequencing (NGS) has decreased, the amount of genomic data has surged globally. However, the cost and complexity of the computational resources required continue to be substantial barriers to leveraging big data. A promising solution to these computational challenges is cloud computing, which provides researchers with the necessary CPUs, memory, storage, and software tools. RESULTS Here, we present Closha 2.0, a cloud computing service that offers a user-friendly platform for analyzing massive genomic datasets. Closha 2.0 is designed to provide a cloud-based environment that enables all genomic researchers, including those with limited or no programming experience, to easily analyze their genomic data. The new 2.0 version of Closha has more user-friendly features than the previous 1.0 version. Firstly, the workbench features a script editor that supports Python, R, and shell script programming, enabling users to write scripts and integrate them into their pipelines. This functionality is particularly useful for downstream analysis. Second, Closha 2.0 runs on containers, which execute each tool in an independent environment. This provides a stable environment and prevents dependency issues and version conflicts among tools. Additionally, users can execute each step of a pipeline individually, allowing them to test applications at each stage and adjust parameters to achieve the desired results. We also updated a high-speed data transmission tool called GBox that facilitates the rapid transfer of large datasets. CONCLUSIONS The analysis pipelines on Closha 2.0 are reproducible, with all analysis parameters and inputs being permanently recorded. Closha 2.0 simplifies multi-step analysis with drag-and-drop functionality and provides a user-friendly interface for genomic scientists to obtain accurate results from NGS data. Closha 2.0 is freely available at https://www.kobic.re.kr/closha2 .
Collapse
Affiliation(s)
- Gunhwan Ko
- Korean Bioinformation Center (KOBIC), KRIBB, 125 Gwahangno, Yuseong-gu, Daejeon, 34141, Korea
| | - Pan-Gyu Kim
- Korean Bioinformation Center (KOBIC), KRIBB, 125 Gwahangno, Yuseong-gu, Daejeon, 34141, Korea
| | - Byung-Ha Yoon
- Korean Bioinformation Center (KOBIC), KRIBB, 125 Gwahangno, Yuseong-gu, Daejeon, 34141, Korea
| | - JaeHee Kim
- Korean Bioinformation Center (KOBIC), KRIBB, 125 Gwahangno, Yuseong-gu, Daejeon, 34141, Korea
| | - Wangho Song
- Korean Bioinformation Center (KOBIC), KRIBB, 125 Gwahangno, Yuseong-gu, Daejeon, 34141, Korea
| | - IkSu Byeon
- Korean Bioinformation Center (KOBIC), KRIBB, 125 Gwahangno, Yuseong-gu, Daejeon, 34141, Korea
| | - JongCheol Yoon
- Korean Bioinformation Center (KOBIC), KRIBB, 125 Gwahangno, Yuseong-gu, Daejeon, 34141, Korea
| | - Byungwook Lee
- Korean Bioinformation Center (KOBIC), KRIBB, 125 Gwahangno, Yuseong-gu, Daejeon, 34141, Korea.
| | - Young-Kuk Kim
- Department of Bio-AI Convergence, Chungnam National University, Daejeon, 34134, Korea.
| |
Collapse
|
5
|
Zhang H, Jiang W, Jiang Y, Xu N, Nong L, Li T, Liu R. Investigating the therapeutic potential of hesperidin targeting CRISP2 in intervertebral disc degeneration and cancer risk mitigation. Front Pharmacol 2024; 15:1447152. [PMID: 39268471 PMCID: PMC11390660 DOI: 10.3389/fphar.2024.1447152] [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: 06/11/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Background Intervertebral disc degeneration (IDD) can lead to disc herniation and spinal instability, sometimes requiring surgical intervention. Currently, estrogen has a potential protective effect on IDD, and estrogen is associated with an increased risk of some cancers, such as breast and endometrial cancer. Therefore, it is important to identify natural compounds that estrogen analogues treat IDD while reducing the risk of tumor development. Objective This study aims to explore a natural metabolic treatment strategy by targeting CRISP2 with the natural compound Hesperidin to mimic the protective effects of estrogen on IDD and reduce the risk of tumor development. Methods Microarray data from healthy volunteers and IDD patients were extracted from the Gene Expression Omnibus (GEO) database, and RNA sequencing and clinical data from various cancer types were analyzed. Differentially expressed genes (DEGs) were identified using the Bioconductor Limma package, followed by principal component analysis, volcano plot, and heatmap visualization. Additionally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, CIBERSORT and ssGSEA immune cell infiltration assessments, survival analysis, metabolite enrichment analysis, and molecular docking were performed. Hesperidin's interaction with CRISP2 was further validated through molecular docking and experimental studies. Results Hesperidin significantly reduced the expression of CRISP2, iNOS, and COX2 in IDD models, decreased reactive oxygen species (ROS) and apoptosis, and diminished inflammatory markers. CIBERSORT and ssGSEA analyses revealed a correlation between CRISP2 and immune cell infiltration. Survival analysis demonstrated that CRISP2 expression levels were associated with patient survival across various cancer types. Hesperidin was found to mimic estrogen's effects on IDD and reduce tumor progression. Cell culture and experimental validation confirmed Hesperidin's protective effects on nucleus pulposus cells (NPCs). Conclusion Hesperidin, as a potential natural metabolic regulator, not only has therapeutic effects on IDD but may also synergize with estrogen therapy to promote spinal health without increasing cancer risk. This study presents a new clinical approach for IDD treatment and lays the foundation for further drug development and experimental research.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
- Department of Orthopedics, Gonghe County Hospital of Traditional Chinese Medicine, Hainan, Qinghai, China
| | - Wei Jiang
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yuqing Jiang
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Nanwei Xu
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Luming Nong
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Tengfei Li
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Ruiping Liu
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
6
|
Rudroff T, Rainio O, Klén R. Leveraging Artificial Intelligence to Optimize Transcranial Direct Current Stimulation for Long COVID Management: A Forward-Looking Perspective. Brain Sci 2024; 14:831. [PMID: 39199522 PMCID: PMC11353063 DOI: 10.3390/brainsci14080831] [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: 07/23/2024] [Revised: 08/12/2024] [Accepted: 08/18/2024] [Indexed: 09/01/2024] Open
Abstract
Long COVID (Coronavirus disease), affecting millions globally, presents unprecedented challenges to healthcare systems due to its complex, multifaceted nature and the lack of effective treatments. This perspective review explores the potential of artificial intelligence (AI)-guided transcranial direct current stimulation (tDCS) as an innovative approach to address the urgent need for effective Long COVID management. The authors examine how AI could optimize tDCS protocols, enhance clinical trial design, and facilitate personalized treatment for the heterogeneous manifestations of Long COVID. Key areas discussed include AI-driven personalization of tDCS parameters based on individual patient characteristics and real-time symptom fluctuations, the use of machine learning for patient stratification, and the development of more sensitive outcome measures in clinical trials. This perspective addresses ethical considerations surrounding data privacy, algorithmic bias, and equitable access to AI-enhanced treatments. It also explores challenges and opportunities for implementing AI-guided tDCS across diverse healthcare settings globally. Future research directions are outlined, including the need for large-scale validation studies and investigations of long-term efficacy and safety. The authors argue that while AI-guided tDCS shows promise for addressing the complex nature of Long COVID, significant technical, ethical, and practical challenges remain. They emphasize the importance of interdisciplinary collaboration, patient-centered approaches, and a commitment to global health equity in realizing the potential of this technology. This perspective article provides a roadmap for researchers, clinicians, and policymakers involved in developing and implementing AI-guided neuromodulation therapies for Long COVID and potentially other neurological and psychiatric conditions.
Collapse
Affiliation(s)
- Thorsten Rudroff
- Turku PET Centre, University of Turku, Turku University Hospital, 20520 Turku, Finland; (O.R.); (R.K.)
| | | | | |
Collapse
|
7
|
Rosa LS, Argolo CO, Nascimento CM, Pimentel AS. Identifying Substructures That Facilitate Compounds to Penetrate the Blood-Brain Barrier via Passive Transport Using Machine Learning Explainer Models. ACS Chem Neurosci 2024; 15:2144-2159. [PMID: 38723285 PMCID: PMC11157485 DOI: 10.1021/acschemneuro.3c00840] [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: 12/26/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 06/06/2024] Open
Abstract
The local interpretable model-agnostic explanation (LIME) method was used to interpret two machine learning models of compounds penetrating the blood-brain barrier. The classification models, Random Forest, ExtraTrees, and Deep Residual Network, were trained and validated using the blood-brain barrier penetration dataset, which shows the penetrability of compounds in the blood-brain barrier. LIME was able to create explanations for such penetrability, highlighting the most important substructures of molecules that affect drug penetration in the barrier. The simple and intuitive outputs prove the applicability of this explainable model to interpreting the permeability of compounds across the blood-brain barrier in terms of molecular features. LIME explanations were filtered with a weight equal to or greater than 0.1 to obtain only the most relevant explanations. The results showed several structures that are important for blood-brain barrier penetration. In general, it was found that some compounds with nitrogenous substructures are more likely to permeate the blood-brain barrier. The application of these structural explanations may help the pharmaceutical industry and potential drug synthesis research groups to synthesize active molecules more rationally.
Collapse
Affiliation(s)
- Lucca
Caiaffa Santos Rosa
- Departamento de Química, Pontifícia Universidade Católica do
Rio de Janeiro, Rio de
Janeiro, RJ 22453-900, Brazil
| | - Caio Oliveira Argolo
- Departamento de Química, Pontifícia Universidade Católica do
Rio de Janeiro, Rio de
Janeiro, RJ 22453-900, Brazil
| | | | - Andre Silva Pimentel
- Departamento de Química, Pontifícia Universidade Católica do
Rio de Janeiro, Rio de
Janeiro, RJ 22453-900, Brazil
| |
Collapse
|
8
|
Wang J, Xue M, Hu Y, Li J, Li Z, Wang Y. Proteomic Insights into Osteoporosis: Unraveling Diagnostic Markers of and Therapeutic Targets for the Metabolic Bone Disease. Biomolecules 2024; 14:554. [PMID: 38785961 PMCID: PMC11118602 DOI: 10.3390/biom14050554] [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: 03/29/2024] [Revised: 04/25/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Osteoporosis (OP), a prevalent skeletal disorder characterized by compromised bone strength and increased susceptibility to fractures, poses a significant public health concern. This review aims to provide a comprehensive analysis of the current state of research in the field, focusing on the application of proteomic techniques to elucidate diagnostic markers and therapeutic targets for OP. The integration of cutting-edge proteomic technologies has enabled the identification and quantification of proteins associated with bone metabolism, leading to a deeper understanding of the molecular mechanisms underlying OP. In this review, we systematically examine recent advancements in proteomic studies related to OP, emphasizing the identification of potential biomarkers for OP diagnosis and the discovery of novel therapeutic targets. Additionally, we discuss the challenges and future directions in the field, highlighting the potential impact of proteomic research in transforming the landscape of OP diagnosis and treatment.
Collapse
Affiliation(s)
- Jihan Wang
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China; (J.W.)
| | - Mengju Xue
- School of Medicine, Xi’an International University, Xi’an 710077, China
| | - Ya Hu
- Department of Medical College, Hunan Polytechnic of Environment and Biology, Hengyang 421000, China
| | - Jingwen Li
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China; (J.W.)
- Research and Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Zhenzhen Li
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China; (J.W.)
- Research and Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Yangyang Wang
- School of Electronics and Information, Northwestern Polytechnical University, Xi’an 710129, China
| |
Collapse
|
9
|
Buch AM, Liston C, Grosenick L. Simple and Scalable Algorithms for Cluster-Aware Precision Medicine. PROCEEDINGS OF MACHINE LEARNING RESEARCH 2024; 238:136-144. [PMID: 39015742 PMCID: PMC11251711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
AI-enabled precision medicine promises a transformational improvement in healthcare outcomes. However, training on biomedical data presents significant challenges as they are often high dimensional, clustered, and of limited sample size. To overcome these challenges, we propose a simple and scalable approach for cluster-aware embedding that combines latent factor methods with a convex clustering penalty in a modular way. Our novel approach overcomes the complexity and limitations of current joint embedding and clustering methods and enables hierarchically clustered principal component analysis (PCA), locally linear embedding (LLE), and canonical correlation analysis (CCA). Through numerical experiments and real-world examples, we demonstrate that our approach outperforms fourteen clustering methods on highly underdetermined problems (e.g., with limited sample size) as well as on large sample datasets. Importantly, our approach does not require the user to choose the desired number of clusters, yields improved model selection if they do, and yields interpretable hierarchically clustered embedding dendrograms. Thus, our approach improves significantly on existing methods for identifying patient subgroups in multiomics and neuroimaging data and enables scalable and interpretable biomarkers for precision medicine.
Collapse
Affiliation(s)
- Amanda M Buch
- Dept. of Psychiatry & BMRI, Weill Cornell Medicine, Cornell University
| | - Conor Liston
- Dept. of Psychiatry & BMRI, Weill Cornell Medicine, Cornell University
| | - Logan Grosenick
- Dept. of Psychiatry & BMRI, Weill Cornell Medicine, Cornell University
| |
Collapse
|
10
|
da Silva Rosa SC, Barzegar Behrooz A, Guedes S, Vitorino R, Ghavami S. Prioritization of genes for translation: a computational approach. Expert Rev Proteomics 2024; 21:125-147. [PMID: 38563427 DOI: 10.1080/14789450.2024.2337004] [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: 05/26/2023] [Accepted: 02/21/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Gene identification for genetic diseases is critical for the development of new diagnostic approaches and personalized treatment options. Prioritization of gene translation is an important consideration in the molecular biology field, allowing researchers to focus on the most promising candidates for further investigation. AREAS COVERED In this paper, we discussed different approaches to prioritize genes for translation, including the use of computational tools and machine learning algorithms, as well as experimental techniques such as knockdown and overexpression studies. We also explored the potential biases and limitations of these approaches and proposed strategies to improve the accuracy and reliability of gene prioritization methods. Although numerous computational methods have been developed for this purpose, there is a need for computational methods that incorporate tissue-specific information to enable more accurate prioritization of candidate genes. Such methods should provide tissue-specific predictions, insights into underlying disease mechanisms, and more accurate prioritization of genes. EXPERT OPINION Using advanced computational tools and machine learning algorithms to prioritize genes, we can identify potential targets for therapeutic intervention of complex diseases. This represents an up-and-coming method for drug development and personalized medicine.
Collapse
Affiliation(s)
- Simone C da Silva Rosa
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Canada
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Canada
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sofia Guedes
- LAQV/REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Rui Vitorino
- LAQV/REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, Aveiro, Portugal
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Canada
- Faculty of Medicine in Zabrze, Academia of Silesia, Katowice, Poland
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
11
|
Colen L, Belderbos R, Kelchtermans S, Leten B. Many are called, few are chosen: the role of science in drug development decisions. JOURNAL OF TECHNOLOGY TRANSFER 2023:1-26. [PMID: 37359814 PMCID: PMC10008718 DOI: 10.1007/s10961-022-09982-6] [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] [Accepted: 11/10/2022] [Indexed: 03/14/2023]
Abstract
Pharmaceutical firms are extremely selective in deciding which patented drug candidates are taken up into clinical development, given the high costs and risks involved. We argue that the scientific base of drug candidates, and who was responsible for that scientific research, are key antecedents of take-up into clinical trials and whether the patent owner ('internal take-up') or another firm ('external take-up') leads the clinical development effort. We hypothesize that patented drug candidates that refer to scientific research are more likely to be taken up in development, and that in-house conducted scientific research is predominantly associated with internal take-up due to the ease of knowledge transfer within the firm. Examining 18,360 drug candidates patented by 136 pharmaceutical firms we find support for these hypotheses. In addition, drug candidates referring to in-house scientific research exhibit a higher probability of eventual drug development success. Our findings underline the importance of a 'rational drug design' approach that explicitly builds on scientific research. The benefits of internal scientific research in clinical development highlight the potential downside of pervasive organizational specialization in the life sciences in either scientific research or clinical development.
Collapse
Affiliation(s)
| | - René Belderbos
- KU Leuven, Leuven, Belgium
- Maastricht University, Maastricht, The Netherlands
- UNU MERIT, Maastricht, The Netherlands
| | | | - Bart Leten
- Hasselt University, Hasselt, Belgium
- KU Leuven, Leuven, Belgium
| |
Collapse
|
12
|
Quan X, Cai W, Xi C, Wang C, Yan L. AIMedGraph: a comprehensive multi-relational knowledge graph for precision medicine. Database (Oxford) 2023; 2023:7059703. [PMID: 36856726 PMCID: PMC9976745 DOI: 10.1093/database/baad006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/01/2023] [Accepted: 02/10/2023] [Indexed: 03/02/2023]
Abstract
The development of high-throughput molecular testing techniques has enabled the large-scale exploration of the underlying molecular causes of diseases and the development of targeted treatment for specific genetic alterations. However, knowledge to interpret the impact of genetic variants on disease or treatment is distributed in different databases, scientific literature studies and clinical guidelines. AIMedGraph was designed to comprehensively collect and interrogate standardized information about genes, genetic alterations and their therapeutic and diagnostic relevance and build a multi-relational, evidence-based knowledge graph. Graph database Neo4j was used to represent precision medicine knowledge as nodes and edges in AIMedGraph. Entities in the current release include 30 340 diseases/phenotypes, 26 140 genes, 187 541 genetic variants, 2821 drugs, 15 125 clinical trials and 797 911 supporting literature studies. Edges in this release cover 621 731 drug interactions, 9279 drug susceptibility impacts, 6330 pharmacogenomics effects, 30 339 variant pathogenicity and 1485 drug adverse reactions. The knowledge graph technique enables hidden knowledge inference and provides insight into potential disease or drug molecular mechanisms. Database URL: http://aimedgraph.tongshugene.net:8201.
Collapse
Affiliation(s)
- Xueping Quan
- Correspondence may also be addressed to Xueping Quan. Tel: +8621-58886662;
| | - Weijing Cai
- Department of Innovative Technology, Shanghai Tongshu Biotechnology Research Institute, No26 and 28, 377 Lane of Shanlian Road, Baoshan District, Shanghai 200444, China
| | - Chenghang Xi
- Department of Artificial Intelligence, Shanghai Tongshu Biotechnology Research Institute, No26 and 28, 377 Lane of Shanlian Road, Baoshan District, Shanghai 200444, China
| | - Chunxiao Wang
- Department of Innovative Technology, Shanghai Tongshu Biotechnology Research Institute, No26 and 28, 377 Lane of Shanlian Road, Baoshan District, Shanghai 200444, China
| | | |
Collapse
|
13
|
Djokovic N, Rahnasto-Rilla M, Lougiakis N, Lahtela-Kakkonen M, Nikolic K. SIRT2i_Predictor: A Machine Learning-Based Tool to Facilitate the Discovery of Novel SIRT2 Inhibitors. Pharmaceuticals (Basel) 2023; 16:ph16010127. [PMID: 36678624 PMCID: PMC9864763 DOI: 10.3390/ph16010127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
A growing body of preclinical evidence recognized selective sirtuin 2 (SIRT2) inhibitors as novel therapeutics for treatment of age-related diseases. However, none of the SIRT2 inhibitors have reached clinical trials yet. Transformative potential of machine learning (ML) in early stages of drug discovery has been witnessed by widespread adoption of these techniques in recent years. Despite great potential, there is a lack of robust and large-scale ML models for discovery of novel SIRT2 inhibitors. In order to support virtual screening (VS), lead optimization, or facilitate the selection of SIRT2 inhibitors for experimental evaluation, a machine-learning-based tool titled SIRT2i_Predictor was developed. The tool was built on a panel of high-quality ML regression and classification-based models for prediction of inhibitor potency and SIRT1-3 isoform selectivity. State-of-the-art ML algorithms were used to train the models on a large and diverse dataset containing 1797 compounds. Benchmarking against structure-based VS protocol indicated comparable coverage of chemical space with great gain in speed. The tool was applied to screen the in-house database of compounds, corroborating the utility in the prioritization of compounds for costly in vitro screening campaigns. The easy-to-use web-based interface makes SIRT2i_Predictor a convenient tool for the wider community. The SIRT2i_Predictor's source code is made available online.
Collapse
Affiliation(s)
- Nemanja Djokovic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
- Correspondence: (N.D.); (K.N.)
| | - Minna Rahnasto-Rilla
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, 70210 Kuopio, Finland
| | - Nikolaos Lougiakis
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece
| | | | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
- Correspondence: (N.D.); (K.N.)
| |
Collapse
|
14
|
Fosse V, Oldoni E, Bietrix F, Budillon A, Daskalopoulos EP, Fratelli M, Gerlach B, Groenen PMA, Hölter SM, Menon JML, Mobasheri A, Osborne N, Ritskes-Hoitinga M, Ryll B, Schmitt E, Ussi A, Andreu AL, McCormack E. Recommendations for robust and reproducible preclinical research in personalised medicine. BMC Med 2023; 21:14. [PMID: 36617553 PMCID: PMC9826728 DOI: 10.1186/s12916-022-02719-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Personalised medicine is a medical model that aims to provide tailor-made prevention and treatment strategies for defined groups of individuals. The concept brings new challenges to the translational step, both in clinical relevance and validity of models. We have developed a set of recommendations aimed at improving the robustness of preclinical methods in translational research for personalised medicine. METHODS These recommendations have been developed following four main steps: (1) a scoping review of the literature with a gap analysis, (2) working sessions with a wide range of experts in the field, (3) a consensus workshop, and (4) preparation of the final set of recommendations. RESULTS Despite the progress in developing innovative and complex preclinical model systems, to date there are fundamental deficits in translational methods that prevent the further development of personalised medicine. The literature review highlighted five main gaps, relating to the relevance of experimental models, quality assessment practices, reporting, regulation, and a gap between preclinical and clinical research. We identified five points of focus for the recommendations, based on the consensus reached during the consultation meetings: (1) clinically relevant translational research, (2) robust model development, (3) transparency and education, (4) revised regulation, and (5) interaction with clinical research and patient engagement. Here, we present a set of 15 recommendations aimed at improving the robustness of preclinical methods in translational research for personalised medicine. CONCLUSIONS Appropriate preclinical models should be an integral contributor to interventional clinical trial success rates, and predictive translational models are a fundamental requirement to realise the dream of personalised medicine. The implementation of these guidelines is ambitious, and it is only through the active involvement of all relevant stakeholders in this field that we will be able to make an impact and effectuate a change which will facilitate improved translation of personalised medicine in the future.
Collapse
Affiliation(s)
- Vibeke Fosse
- Department of Clinical Science, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway.
| | - Emanuela Oldoni
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, The Netherlands
| | - Florence Bietrix
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, The Netherlands
| | - Alfredo Budillon
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | | | - Maddalena Fratelli
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Björn Gerlach
- PAASP GmbH, Guarantors of EQIPD e.V., Central Institute for Mental Health in Mannheim, Mannheim, Germany
| | | | | | - Julia M L Menon
- Preclinicaltrials.eu, Netherlands Heart Institute, Utrecht, The Netherlands
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, 90570, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406, Vilnius, Lithuania
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 508, GA, Utrecht, The Netherlands
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000, Liège, Belgium
| | | | - Merel Ritskes-Hoitinga
- Department of Population Health Sciences, IRAS, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Department of Clinical Medicine, AUGUST, Aarhus University, Aarhus, Denmark
| | - Bettina Ryll
- Melanoma Patient Network Europe, Uppsala, Sweden
| | - Elmar Schmitt
- Global Regulatory Oncology, Merck Healthcare KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Anton Ussi
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, The Netherlands
| | - Antonio L Andreu
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, The Netherlands
| | - Emmet McCormack
- Department of Clinical Science, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
- Department of Clinical Science, Centre for Pharmacy, The University of Bergen, Bergen, Norway
| |
Collapse
|
15
|
Abstract
Liver cancer, mainly hepatocellular carcinoma (HCC), remains a major cause of cancer-related death worldwide. With the global epidemic of obesity, the major HCC etiologies have been dynamically shifting from viral to metabolic liver diseases. This change has made HCC prevention difficult with increasingly elusive at-risk populations as rational target for preventive interventions. Besides ongoing efforts to reduce obesity and metabolic disorders, chemoprevention in patients who already have metabolic liver diseases may have a significant impact on the poor HCC prognosis. Hepatitis B- and hepatitis C-related HCC incidences have been substantially reduced by the new antivirals, but HCC risk can persist over a decade even after successful viral treatment, highlighting the need for HCC-preventive measures also in these patients. Experimental and retrospective studies have suggested potential utility of generic agents such as lipophilic statins and aspirin for HCC chemoprevention given their well-characterized safety profile, although anticipated efficacy may be modest. In this review, we overview recent clinical and translational studies of generic agents in the context of HCC chemoprevention under the contemporary HCC etiologies. We also discuss newly emerging approaches to overcome the challenges in clinical testing of the agents to facilitate their clinical translation.
Collapse
Affiliation(s)
- Fahmida Rasha
- Liver Tumor Translational Research Program; Simmons Comprehensive Cancer Center; Division of Digestive and Liver Diseases; Department of Internal Medicine; University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Subhojit Paul
- Liver Tumor Translational Research Program; Simmons Comprehensive Cancer Center; Division of Digestive and Liver Diseases; Department of Internal Medicine; University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tracey G Simon
- Liver Center, Division of Gastroenterology, Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yujin Hoshida
- Liver Tumor Translational Research Program; Simmons Comprehensive Cancer Center; Division of Digestive and Liver Diseases; Department of Internal Medicine; University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
16
|
Pavel A, Saarimäki LA, Möbus L, Federico A, Serra A, Greco D. The potential of a data centred approach & knowledge graph data representation in chemical safety and drug design. Comput Struct Biotechnol J 2022; 20:4837-4849. [PMID: 36147662 PMCID: PMC9464643 DOI: 10.1016/j.csbj.2022.08.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 11/20/2022] Open
Abstract
Big Data pervades nearly all areas of life sciences, yet the analysis of large integrated data sets remains a major challenge. Moreover, the field of life sciences is highly fragmented and, consequently, so is its data, knowledge, and standards. This, in turn, makes integrated data analysis and knowledge gathering across sub-fields a demanding task. At the same time, the integration of various research angles and data types is crucial for modelling the complexity of organisms and biological processes in a holistic manner. This is especially valid in the context of drug development and chemical safety assessment where computational methods can provide solutions for the urgent need of fast, effective, and sustainable approaches. At the same time, such computational methods require the development of methodologies suitable for an integrated and data centred Big Data view. Here we discuss Knowledge Graphs (KG) as a solution to a data centred analysis approach for drug and chemical development and safety assessment. KGs are knowledge bases, data analysis engines, and knowledge discovery systems all in one, allowing them to be used from simple data retrieval, over meta-analysis to complex predictive and knowledge discovery systems. Therefore, KGs have immense potential to advance the data centred approach, the re-usability, and informativity of data. Furthermore, they can improve the power of analysis, and the complexity of modelled processes, all while providing knowledge in a natively human understandable network data model.
Collapse
Affiliation(s)
- Alisa Pavel
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Laura A Saarimäki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Lena Möbus
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland.,Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
17
|
Abstract
Three-dimensional protein structural data at the molecular level are pivotal for successful precision medicine. Such data are crucial not only for discovering drugs that act to block the active site of the target mutant protein but also for clarifying to the patient and the clinician how the mutations harbored by the patient work. The relative paucity of structural data reflects their cost, challenges in their interpretation, and lack of clinical guidelines for their utilization. Rapid technological advancements in experimental high-resolution structural determination increasingly generate structures. Computationally, modeling algorithms, including molecular dynamics simulations, are becoming more powerful, as are compute-intensive hardware, particularly graphics processing units, overlapping with the inception of the exascale era. Accessible, freely available, and detailed structural and dynamical data can be merged with big data to powerfully transform personalized pharmacology. Here we review protein and emerging genome high-resolution data, along with means, applications, and examples underscoring their usefulness in precision medicine. Expected final online publication date for the Annual Review of Biomedical Data Science, Volume 5 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, Maryland, USA; .,Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, Maryland, USA;
| | - Guy Nir
- Department of Biochemistry and Molecular Biology, Department of Neuroscience, Cell Biology and Anatomy, and Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, Maryland, USA;
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
18
|
Big Data Precision Marketing Approach under IoT Cloud Platform Information Mining. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:4828108. [PMID: 35069719 PMCID: PMC8769856 DOI: 10.1155/2022/4828108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 02/03/2023]
Abstract
In this article, an in-depth study and analysis of the precision marketing approach are carried out by building an IoT cloud platform and then using the technology of big data information mining. The cloud platform uses the MySQL database combined with the MongoDB database to store the cloud platform data to ensure the correct storage of data as well as to improve the access speed of data. The storage method of IoT temporal data is optimized, and the way of storing data in time slots is used to improve the efficiency of reading large amounts of data. For the scalability of the IoT data storage system, a MongoDB database clustering scheme is designed to ensure the scalability of data storage and disaster recovery capability. The relevant theories of big data marketing are reviewed and analyzed; secondly, based on the relevant theories, combined with the author’s work experience and relevant information, a comprehensive analysis and research on the current situation of big data marketing are conducted, focusing on its macro-, micro-, and industry environment. The service model combines the types of user needs, encapsulates the resources obtained by the alliance through data mining for service products, and publishes and delivers them in the form of data products. From the perspective of the development of the telecommunications industry, in terms of technology, the telecommunications industry has seen the development trend of mobile replacing fixed networks and triple play. The development of emerging technologies represented by the Internet of Things and cloud computing has also led to technological changes in the telecommunications industry. Operators are facing new development opportunities and challenges. It also divides the service mode into self-service and consulting service mode according to the different degrees of users’ cognition and understanding of the service, as well as proposes standardized data mining service guarantee from two aspects: after-sales service and operation supervision. A customized data mining service is a kind of data mining service for users’ personalized needs. And the intelligent data mining service guarantee is proposed from two aspects of multicase experience integration and group intelligence. In the empirical research part, the big data alliance in Big Data Industry Alliance, which provides data mining service as the main business, is selected as the research object, and the data mining service model of the big data alliance proposed in this article is applied to the actual alliance to verify the scientific and rationality of the data mining service model and improve the data mining service model management system.
Collapse
|
19
|
Shahzadi S, Yasir M, Aftab B, Babar S, Hassan M. Exploration of Protein Aggregations in Parkinson's Disease Through Computational Approaches and Big Data Analytics. Methods Mol Biol 2022; 2340:449-467. [PMID: 35167085 DOI: 10.1007/978-1-0716-1546-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Protein aggregation has been implicated in numerous neurodegenerative disorders whose etiologies are poorly understood, and for which there are no effective treatments. Here we show that the computational approaches may help us to better understand the basics of Parkinson's disease (PD). The high-resolution structural, dynamical, and mechanistic insights delivered by computational studies of protein aggregation have a unique potential to enable the rational manipulation of oligomer formation. Additionally, big data and machine learning methods may provide valuable insights to better understand the nature of proteins involved in PD and their aggregative behavior for the betterment of PD treatment.
Collapse
Affiliation(s)
- Saba Shahzadi
- Institute of Molecular Sciences and Bioinformatics, Lahore, Pakistan
| | - Muhammad Yasir
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Bisma Aftab
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Sumbal Babar
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Mubashir Hassan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan.
- Battelle Center for Mathematical Medicine, Nationwide Children Hospital & Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
20
|
Rando HM, Wellhausen N, Ghosh S, Lee AJ, Dattoli AA, Hu F, Byrd JB, Rafizadeh DN, Lordan R, Qi Y, Sun Y, Brueffer C, Field JM, Ben Guebila M, Jadavji NM, Skelly AN, Ramsundar B, Wang J, Goel RR, Park Y, Boca SM, Gitter A, Greene CS. Identification and Development of Therapeutics for COVID-19. mSystems 2021; 6:e0023321. [PMID: 34726496 PMCID: PMC8562484 DOI: 10.1128/msystems.00233-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
After emerging in China in late 2019, the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spread worldwide, and as of mid-2021, it remains a significant threat globally. Only a few coronaviruses are known to infect humans, and only two cause infections similar in severity to SARS-CoV-2: Severe acute respiratory syndrome-related coronavirus, a species closely related to SARS-CoV-2 that emerged in 2002, and Middle East respiratory syndrome-related coronavirus, which emerged in 2012. Unlike the current pandemic, previous epidemics were controlled rapidly through public health measures, but the body of research investigating severe acute respiratory syndrome and Middle East respiratory syndrome has proven valuable for identifying approaches to treating and preventing novel coronavirus disease 2019 (COVID-19). Building on this research, the medical and scientific communities have responded rapidly to the COVID-19 crisis and identified many candidate therapeutics. The approaches used to identify candidates fall into four main categories: adaptation of clinical approaches to diseases with related pathologies, adaptation based on virological properties, adaptation based on host response, and data-driven identification (ID) of candidates based on physical properties or on pharmacological compendia. To date, a small number of therapeutics have already been authorized by regulatory agencies such as the Food and Drug Administration (FDA), while most remain under investigation. The scale of the COVID-19 crisis offers a rare opportunity to collect data on the effects of candidate therapeutics. This information provides insight not only into the management of coronavirus diseases but also into the relative success of different approaches to identifying candidate therapeutics against an emerging disease. IMPORTANCE The COVID-19 pandemic is a rapidly evolving crisis. With the worldwide scientific community shifting focus onto the SARS-CoV-2 virus and COVID-19, a large number of possible pharmaceutical approaches for treatment and prevention have been proposed. What was known about each of these potential interventions evolved rapidly throughout 2020 and 2021. This fast-paced area of research provides important insight into how the ongoing pandemic can be managed and also demonstrates the power of interdisciplinary collaboration to rapidly understand a virus and match its characteristics with existing or novel pharmaceuticals. As illustrated by the continued threat of viral epidemics during the current millennium, a rapid and strategic response to emerging viral threats can save lives. In this review, we explore how different modes of identifying candidate therapeutics have borne out during COVID-19.
Collapse
Affiliation(s)
- Halie M. Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Soumita Ghosh
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alexandra J. Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna Ada Dattoli
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fengling Hu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James Brian Byrd
- University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Diane N. Rafizadeh
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yanjun Qi
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
| | - Yuchen Sun
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
| | | | - Jeffrey M. Field
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marouen Ben Guebila
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Nafisa M. Jadavji
- Biomedical Science, Midwestern University, Glendale, Arizona, USA
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Ashwin N. Skelly
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Jinhui Wang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rishi Raj Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - YoSon Park
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - COVID-19 Review Consortium
BansalVikasBartonJohn P.BocaSimina M.BoerckelJoel D.BruefferChristianByrdJames BrianCaponeStephenDasShiktaDattoliAnna AdaDziakJohn J.FieldJeffrey M.GhoshSoumitaGitterAnthonyGoelRishi RajGreeneCasey S.GuebilaMarouen BenHimmelsteinDaniel S.HuFenglingJadavjiNafisa M.KamilJeremy P.KnyazevSergeyKollaLikhithaLeeAlexandra J.LordanRonanLubianaTiagoLukanTemitayoMacLeanAdam L.MaiDavidMangulSergheiManheimDavidMcGowanLucy D’AgostinoNaikAmrutaParkYoSonPerrinDimitriQiYanjunRafizadehDiane N.RamsundarBharathRandoHalie M.RaySandipanRobsonMichael P.RubinettiVincentSellElizabethShinholsterLamonicaSkellyAshwin N.SunYuchenSunYushaSzetoGregory L.VelazquezRyanWangJinhuiWellhausenNils
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- University of Michigan School of Medicine, Ann Arbor, Michigan, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
- Biomedical Science, Midwestern University, Glendale, Arizona, USA
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- The DeepChem Project
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
- Early Biometrics & Statistical Innovation, Data Science & Artificial Intelligence, R & D, AstraZeneca, Gaithersburg, Maryland, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| | - Simina M. Boca
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
- Early Biometrics & Statistical Innovation, Data Science & Artificial Intelligence, R & D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Casey S. Greene
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Uncovering Quercetin’s Effects against Influenza A Virus Using Network Pharmacology and Molecular Docking. Processes (Basel) 2021. [DOI: 10.3390/pr9091627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
(1) Background: Re-emerging influenza threats continue to challenge medical and public health systems. Quercetin is a ubiquitous flavonoid found in food and is recognized to possess antioxidant, anti-inflammatory, antiviral, and anticancer activities. (2) Methods: To elucidate the targets and mechanisms underlying the action of quercetin as a therapeutic agent for influenza, network pharmacology and molecular docking were employed. Biological targets of quercetin and target genes associated with influenza were retrieved from public databases. Compound–disease target (C-D) networks were constructed, and targets were further analyzed using KEGG pathway analysis. Potent target genes were retrieved from the compound–disease–pathway (C-D-P) and protein–protein interaction (PPI) networks. The binding affinities between quercetin and the targets were identified using molecular docking. (3) Results: The pathway study revealed that quercetin-associated influenza targets were mainly involved in viral diseases, inflammation-associated pathways, and cancer. Four targets, MAPK1, NFKB1, RELA, and TP53, were identified to be involved in the inhibitory effects of quercetin on influenza. Using the molecular docking method, we evaluated the binding affinity of each ligand (quercetin)–target and discovered that quercetin and MAPK1 showed the strongest calculated binding energy among the four ligand–target complexes. (4) Conclusion: These findings identified potential targets of quercetin and suggest quercetin as a potential drug for influenza treatment.
Collapse
|
22
|
The Assessment of Big Data Adoption Readiness with a Technology–Organization–Environment Framework: A Perspective towards Healthcare Employees. SUSTAINABILITY 2021. [DOI: 10.3390/su13158379] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Big data is rapidly being seen as a new frontier for improving organizational performance. However, it is still in its early phases of implementation in developing countries’ healthcare organizations. As data-driven insights become critical competitive advantages, it is critical to ascertain which elements influence an organization’s decision to adopt big data. The aim of this study is to propose and empirically test a theoretical framework based on technology–organization–environment (TOE) factors to identify the level of readiness of big data adoption in developing countries’ healthcare organizations. The framework empirically tested 302 Malaysian healthcare employees. The structural equation modeling was used to analyze the collected data. The results of the study demonstrated that technology, organization, and environment factors can significantly contribute towards big data adoption in healthcare organizations. However, the complexity of technology factors has shown less support for the notion. For technology practitioners, this study showed how to enhance big data adoption in healthcare organizations through TOE factors.
Collapse
|
23
|
Mavridou D, Psatha K, Aivaliotis M. Proteomics and Drug Repurposing in CLL towards Precision Medicine. Cancers (Basel) 2021; 13:cancers13143391. [PMID: 34298607 PMCID: PMC8303629 DOI: 10.3390/cancers13143391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Despite continued efforts, the current status of knowledge in CLL molecular pathobiology, diagnosis, prognosis and treatment remains elusive and imprecise. Proteomics approaches combined with advanced bioinformatics and drug repurposing promise to shed light on the complex proteome heterogeneity of CLL patients and mitigate, improve, or even eliminate the knowledge stagnation. In relation to this concept, this review presents a brief overview of all the available proteomics and drug repurposing studies in CLL and suggests the way such studies can be exploited to find effective therapeutic options combined with drug repurposing strategies to adopt and accost a more “precision medicine” spectrum. Abstract CLL is a hematological malignancy considered as the most frequent lymphoproliferative disease in the western world. It is characterized by high molecular heterogeneity and despite the available therapeutic options, there are many patient subgroups showing the insufficient effectiveness of disease treatment. The challenge is to investigate the individual molecular characteristics and heterogeneity of these patients. Proteomics analysis is a powerful approach that monitors the constant state of flux operators of genetic information and can unravel the proteome heterogeneity and rewiring into protein pathways in CLL patients. This review essences all the available proteomics studies in CLL and suggests the way these studies can be exploited to find effective therapeutic options combined with drug repurposing approaches. Drug repurposing utilizes all the existing knowledge of the safety and efficacy of FDA-approved or investigational drugs and anticipates drug alignment to crucial CLL therapeutic targets, leading to a better disease outcome. The drug repurposing studies in CLL are also discussed in this review. The next goal involves the integration of proteomics-based drug repurposing in precision medicine, as well as the application of this procedure into clinical practice to predict the most appropriate drugs combination that could ensure therapy and the long-term survival of each CLL patient.
Collapse
Affiliation(s)
- Dimitra Mavridou
- Laboratory of Biochemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece;
- Functional Proteomics and Systems Biology (FunPATh)—Center for Interdisciplinary Research and Innovation (CIRI-AUTH), GR-57001 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Konstantina Psatha
- Laboratory of Biochemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece;
- Functional Proteomics and Systems Biology (FunPATh)—Center for Interdisciplinary Research and Innovation (CIRI-AUTH), GR-57001 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, GR-70013 Heraklion, Greece
- Correspondence: (K.P.); (M.A.)
| | - Michalis Aivaliotis
- Laboratory of Biochemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece;
- Functional Proteomics and Systems Biology (FunPATh)—Center for Interdisciplinary Research and Innovation (CIRI-AUTH), GR-57001 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, GR-70013 Heraklion, Greece
- Correspondence: (K.P.); (M.A.)
| |
Collapse
|
24
|
Tripathi MK, Nath A, Singh TP, Ethayathulla AS, Kaur P. Evolving scenario of big data and Artificial Intelligence (AI) in drug discovery. Mol Divers 2021; 25:1439-1460. [PMID: 34159484 PMCID: PMC8219515 DOI: 10.1007/s11030-021-10256-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/14/2021] [Indexed: 12/24/2022]
Abstract
The accumulation of massive data in the plethora of Cheminformatics databases has made the role of big data and artificial intelligence (AI) indispensable in drug design. This has necessitated the development of newer algorithms and architectures to mine these databases and fulfil the specific needs of various drug discovery processes such as virtual drug screening, de novo molecule design and discovery in this big data era. The development of deep learning neural networks and their variants with the corresponding increase in chemical data has resulted in a paradigm shift in information mining pertaining to the chemical space. The present review summarizes the role of big data and AI techniques currently being implemented to satisfy the ever-increasing research demands in drug discovery pipelines.
Collapse
Affiliation(s)
- Manish Kumar Tripathi
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Abhigyan Nath
- Department of Biochemistry, Pt. Jawahar Lal Nehru Memorial Medical College, Raipur, 492001, India
| | - Tej P Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - A S Ethayathulla
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
25
|
Karamperis K, Tsoumpeli MT, Kounelis F, Koromina M, Mitropoulou C, Moutinho C, Patrinos GP. Genome-based therapeutic interventions for β-type hemoglobinopathies. Hum Genomics 2021; 15:32. [PMID: 34090531 PMCID: PMC8178887 DOI: 10.1186/s40246-021-00329-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022] Open
Abstract
For decades, various strategies have been proposed to solve the enigma of hemoglobinopathies, especially severe cases. However, most of them seem to be lagging in terms of effectiveness and safety. So far, the most prevalent and promising treatment options for patients with β-types hemoglobinopathies, among others, predominantly include drug treatment and gene therapy. Despite the significant improvements of such interventions to the patient's quality of life, a variable response has been demonstrated among different groups of patients and populations. This is essentially due to the complexity of the disease and other genetic factors. In recent years, a more in-depth understanding of the molecular basis of the β-type hemoglobinopathies has led to significant upgrades to the current technologies, as well as the addition of new ones attempting to elucidate these barriers. Therefore, the purpose of this article is to shed light on pharmacogenomics, gene addition, and genome editing technologies, and consequently, their potential use as direct and indirect genome-based interventions, in different strategies, referring to drug and gene therapy. Furthermore, all the latest progress, updates, and scientific achievements for patients with β-type hemoglobinopathies will be described in detail.
Collapse
Affiliation(s)
- Kariofyllis Karamperis
- Department of Pharmacy, School of Health Sciences, Laboratory of Pharmacogenomics and Individualized Therapy, University of Patras, Patras, Greece
- The Golden Helix Foundation, London, UK
| | - Maria T Tsoumpeli
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Fotios Kounelis
- Department of Computing, Group of Large-Scale Data & Systems, Imperial College London, London, UK
| | - Maria Koromina
- Department of Pharmacy, School of Health Sciences, Laboratory of Pharmacogenomics and Individualized Therapy, University of Patras, Patras, Greece
| | | | - Catia Moutinho
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - George P Patrinos
- Department of Pharmacy, School of Health Sciences, Laboratory of Pharmacogenomics and Individualized Therapy, University of Patras, Patras, Greece.
- College of Medicine and Health Sciences, Department of Pathology, United Arab Emirates University, Al-Ain, United Arab Emirates.
- Zayed Center of Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
| |
Collapse
|
26
|
Boniolo F, Dorigatti E, Ohnmacht AJ, Saur D, Schubert B, Menden MP. Artificial intelligence in early drug discovery enabling precision medicine. Expert Opin Drug Discov 2021; 16:991-1007. [PMID: 34075855 DOI: 10.1080/17460441.2021.1918096] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Precision medicine is the concept of treating diseases based on environmental factors, lifestyles, and molecular profiles of patients. This approach has been found to increase success rates of clinical trials and accelerate drug approvals. However, current precision medicine applications in early drug discovery use only a handful of molecular biomarkers to make decisions, whilst clinics gear up to capture the full molecular landscape of patients in the near future. This deep multi-omics characterization demands new analysis strategies to identify appropriate treatment regimens, which we envision will be pioneered by artificial intelligence.Areas covered: In this review, the authors discuss the current state of drug discovery in precision medicine and present our vision of how artificial intelligence will impact biomarker discovery and drug design.Expert opinion: Precision medicine is expected to revolutionize modern medicine; however, its traditional form is focusing on a few biomarkers, thus not equipped to leverage the full power of molecular landscapes. For learning how the development of drugs can be tailored to the heterogeneity of patients across their molecular profiles, artificial intelligence algorithms are the next frontier in precision medicine and will enable a fully personalized approach in drug design, and thus ultimately impacting clinical practice.
Collapse
Affiliation(s)
- Fabio Boniolo
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,School of Medicine, Chair of Translational Cancer Research and Institute for Experimental Cancer Therapy, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Emilio Dorigatti
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Statistical Learning and Data Science, Department of Statistics, Ludwig Maximilian Universität München, Munich, Germany
| | - Alexander J Ohnmacht
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Department of Biology, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Dieter Saur
- School of Medicine, Chair of Translational Cancer Research and Institute for Experimental Cancer Therapy, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Benjamin Schubert
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Department of Mathematics, Technical University of Munich, Garching, Germany
| | - Michael P Menden
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Department of Biology, Ludwig-Maximilians University Munich, Martinsried, Germany.,German Centre for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
27
|
Kropiwnicki E, Evangelista JE, Stein DJ, Clarke DJB, Lachmann A, Kuleshov MV, Jeon M, Jagodnik KM, Ma’ayan A. Drugmonizome and Drugmonizome-ML: integration and abstraction of small molecule attributes for drug enrichment analysis and machine learning. Database (Oxford) 2021; 2021:baab017. [PMID: 33787872 PMCID: PMC8011435 DOI: 10.1093/database/baab017] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/11/2021] [Accepted: 03/19/2021] [Indexed: 12/15/2022]
Abstract
Understanding the underlying molecular and structural similarities between seemingly heterogeneous sets of drugs can aid in identifying drug repurposing opportunities and assist in the discovery of novel properties of preclinical small molecules. A wealth of information about drug and small molecule structure, targets, indications and side effects; induced gene expression signatures; and other attributes are publicly available through web-based tools, databases and repositories. By processing, abstracting and aggregating information from these resources into drug set libraries, knowledge about novel properties of drugs and small molecules can be systematically imputed with machine learning. In addition, drug set libraries can be used as the underlying database for drug set enrichment analysis. Here, we present Drugmonizome, a database with a search engine for querying annotated sets of drugs and small molecules for performing drug set enrichment analysis. Utilizing the data within Drugmonizome, we also developed Drugmonizome-ML. Drugmonizome-ML enables users to construct customized machine learning pipelines using the drug set libraries from Drugmonizome. To demonstrate the utility of Drugmonizome, drug sets from 12 independent SARS-CoV-2 in vitro screens were subjected to consensus enrichment analysis. Despite the low overlap among these 12 independent in vitro screens, we identified common biological processes critical for blocking viral replication. To demonstrate Drugmonizome-ML, we constructed a machine learning pipeline to predict whether approved and preclinical drugs may induce peripheral neuropathy as a potential side effect. Overall, the Drugmonizome and Drugmonizome-ML resources provide rich and diverse knowledge about drugs and small molecules for direct systems pharmacology applications. Database URL: https://maayanlab.cloud/drugmonizome/.
Collapse
Affiliation(s)
- Eryk Kropiwnicki
- Department of Pharmacological Sciences; Mount Sinai Center for Bioinformatics; Big Data to Knowledge, Library of Integrated Network-Based Cellular Signatures, Data Coordination and Integration Center (BD2K-LINCS DCIC); Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG); Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - John E Evangelista
- Department of Pharmacological Sciences; Mount Sinai Center for Bioinformatics; Big Data to Knowledge, Library of Integrated Network-Based Cellular Signatures, Data Coordination and Integration Center (BD2K-LINCS DCIC); Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG); Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Daniel J Stein
- Department of Pharmacological Sciences; Mount Sinai Center for Bioinformatics; Big Data to Knowledge, Library of Integrated Network-Based Cellular Signatures, Data Coordination and Integration Center (BD2K-LINCS DCIC); Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG); Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Daniel J B Clarke
- Department of Pharmacological Sciences; Mount Sinai Center for Bioinformatics; Big Data to Knowledge, Library of Integrated Network-Based Cellular Signatures, Data Coordination and Integration Center (BD2K-LINCS DCIC); Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG); Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Alexander Lachmann
- Department of Pharmacological Sciences; Mount Sinai Center for Bioinformatics; Big Data to Knowledge, Library of Integrated Network-Based Cellular Signatures, Data Coordination and Integration Center (BD2K-LINCS DCIC); Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG); Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Maxim V Kuleshov
- Department of Pharmacological Sciences; Mount Sinai Center for Bioinformatics; Big Data to Knowledge, Library of Integrated Network-Based Cellular Signatures, Data Coordination and Integration Center (BD2K-LINCS DCIC); Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG); Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Minji Jeon
- Department of Pharmacological Sciences; Mount Sinai Center for Bioinformatics; Big Data to Knowledge, Library of Integrated Network-Based Cellular Signatures, Data Coordination and Integration Center (BD2K-LINCS DCIC); Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG); Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Kathleen M Jagodnik
- Department of Pharmacological Sciences; Mount Sinai Center for Bioinformatics; Big Data to Knowledge, Library of Integrated Network-Based Cellular Signatures, Data Coordination and Integration Center (BD2K-LINCS DCIC); Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG); Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Avi Ma’ayan
- Department of Pharmacological Sciences; Mount Sinai Center for Bioinformatics; Big Data to Knowledge, Library of Integrated Network-Based Cellular Signatures, Data Coordination and Integration Center (BD2K-LINCS DCIC); Knowledge Management Center for Illuminating the Druggable Genome (KMC-IDG); Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| |
Collapse
|
28
|
McComb M, Bies R, Ramanathan M. Machine learning in pharmacometrics: Opportunities and challenges. Br J Clin Pharmacol 2021; 88:1482-1499. [PMID: 33634893 DOI: 10.1111/bcp.14801] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
The explosive growth in medical devices, imaging and diagnostics, computing, and communication and information technologies in drug development and healthcare has created an ever-expanding data landscape that the pharmacometrics (PMX) research community must now traverse. The tools of machine learning (ML) have emerged as a powerful computational approach in other data-rich disciplines but its effective utilization in the pharmaceutical sciences and PMX modelling is in its infancy. ML-based methods can complement PMX modelling by enabling the information in diverse sources of big data, e.g. population-based public databases and disease-specific clinical registries, to be harnessed because they are capable of efficiently identifying salient variables associated with outcomes and delineating their interdependencies. ML algorithms are computationally efficient, have strong predictive capabilities and can enable learning in the big data setting. ML algorithms can be viewed as providing a computational bridge from big data to complement PMX modelling. This review provides an overview of the strengths and weaknesses of ML approaches vis-à-vis population methods, assesses current research into ML applications in the pharmaceutical sciences and provides perspective for potential opportunities and strategies for the successful integration and utilization of ML in PMX.
Collapse
Affiliation(s)
- Mason McComb
- Department of Pharmaceutical Sciences, University at Buffalo, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Robert Bies
- Department of Pharmaceutical Sciences, University at Buffalo, University at Buffalo, State University of New York, Buffalo, NY, USA.,Institute for Computational Data Science, University at Buffalo, NY, USA
| | - Murali Ramanathan
- Department of Pharmaceutical Sciences, University at Buffalo, University at Buffalo, State University of New York, Buffalo, NY, USA.,Department of Neurology, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
29
|
Friedrichs M. BioDWH2: an automated graph-based data warehouse and mapping tool. J Integr Bioinform 2021; 18:167-176. [PMID: 33618440 PMCID: PMC8238471 DOI: 10.1515/jib-2020-0033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 12/28/2022] Open
Abstract
Data integration plays a vital role in scientific research. In biomedical research, the OMICS fields have shown the need for larger datasets, like proteomics, pharmacogenomics, and newer fields like foodomics. As research projects require multiple data sources, mapping between these sources becomes necessary. Utilized workflow systems and integration tools therefore need to process large amounts of heterogeneous data formats, check for data source updates, and find suitable mapping methods to cross-reference entities from different databases. This article presents BioDWH2, an open-source, graph-based data warehouse and mapping tool, capable of helping researchers with these issues. A workspace centered approach allows project-specific data source selections and Neo4j or GraphQL server tools enable quick access to the database for analysis. The BioDWH2 tools are available to the scientific community at https://github.com/BioDWH2.
Collapse
Affiliation(s)
- Marcel Friedrichs
- Bielefeld University, Faculty of Technology, Bioinformatics / Medical Informatics Department, Bielefeld, Germany
| |
Collapse
|
30
|
Assadi M, Jokar N, Ghasemi M, Nabipour I, Gholamrezanezhad A, Ahmadzadehfar H. Precision Medicine Approach in Prostate Cancer. Curr Pharm Des 2021; 26:3783-3798. [PMID: 32067601 DOI: 10.2174/1381612826666200218104921] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 02/12/2020] [Indexed: 12/19/2022]
Abstract
Prostate cancer is the most prevalent type of cancer and the second cause of death in men worldwide. Various diagnostic and treatment procedures are available for this type of malignancy, but High-grade or locally advanced prostate cancers showed the potential to develop to lethal phase that can be causing dead. Therefore, new approaches are needed to prolong patients' survival and to improve their quality of life. Precision medicine is a novel emerging field that plays an essential role in identifying new sub-classifications of diseases and in providing guidance in treatment that is based on individual multi-omics data. Multi-omics approaches include the use of genomics, transcriptomics, proteomics, metabolomics, epigenomics and phenomics data to unravel the complexity of a disease-associated biological network, to predict prognostic biomarkers, and to identify new targeted drugs for individual cancer patients. We review the impact of multi-omics data in the framework of systems biology in the era of precision medicine, emphasising the combination of molecular imaging modalities with highthroughput techniques and the new treatments that target metabolic pathways involved in prostate cancer.
Collapse
Affiliation(s)
- Majid Assadi
- The Persian Gulf Nuclear Medicine Research Center, Department of Molecular Imaging and Radionuclide Therapy (MIRT), Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Narges Jokar
- The Persian Gulf Nuclear Medicine Research Center, Department of Molecular Imaging and Radionuclide Therapy (MIRT), Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mojtaba Ghasemi
- Laboratory of Computational Biotechnology and Bioinformatics (CBB), Department of Plant Breeding and Biotechnology (PBB), Faculty of Agriculture, University of Zabol, Zabol, Iran
| | - Iraj Nabipour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ali Gholamrezanezhad
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo Street, Los Angeles, CA 90033, United States
| | | |
Collapse
|
31
|
Moustafa K. Make good use of big data: A home for everyone. CITIES (LONDON, ENGLAND) 2020; 107:102903. [PMID: 32863523 PMCID: PMC7444630 DOI: 10.1016/j.cities.2020.102903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/11/2020] [Indexed: 05/13/2023]
Abstract
The ongoing COVID-19 pandemic should teach us some lessons at health, environmental and human levels toward more fairness, human cohesion and environmental sustainability. At a health level, the pandemic raises the importance of housing for everyone particularly vulnerable and homeless people to protect them from the disease and against other similar airborne pandemics. Here, I propose to make good use of big data along with 3D construction printers to construct houses and solve some major and pressing housing needs worldwide. Big data can be used to determine how many people do need accommodation and 3D construction printers to build houses accordingly and swiftly. The combination of such facilities- big data and 3D printers- can help solve global housing crises more efficiently than traditional and unguided construction plans, particularly under environmental and major health crises where health and housing are tightly interrelated.
Collapse
|
32
|
Tuerkova A, Zdrazil B. A ligand-based computational drug repurposing pipeline using KNIME and Programmatic Data Access: case studies for rare diseases and COVID-19. J Cheminform 2020; 12:71. [PMID: 33250934 PMCID: PMC7686838 DOI: 10.1186/s13321-020-00474-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/09/2020] [Indexed: 01/01/2023] Open
Abstract
Biomedical information mining is increasingly recognized as a promising technique to accelerate drug discovery and development. Especially, integrative approaches which mine data from several (open) data sources have become more attractive with the increasing possibilities to programmatically access data through Application Programming Interfaces (APIs). The use of open data in conjunction with free, platform-independent analytic tools provides the additional advantage of flexibility, re-usability, and transparency. Here, we present a strategy for performing ligand-based in silico drug repurposing with the analytics platform KNIME. We demonstrate the usefulness of the developed workflow on the basis of two different use cases: a rare disease (here: Glucose Transporter Type 1 (GLUT-1) deficiency), and a new disease (here: COVID 19). The workflow includes a targeted download of data through web services, data curation, detection of enriched structural patterns, as well as substructure searches in DrugBank and a recently deposited data set of antiviral drugs provided by Chemical Abstracts Service. Developed workflows, tutorials with detailed step-by-step instructions, and the information gained by the analysis of data for GLUT-1 deficiency syndrome and COVID-19 are made freely available to the scientific community. The provided framework can be reused by researchers for other in silico drug repurposing projects, and it should serve as a valuable teaching resource for conveying integrative data mining strategies.
Collapse
Affiliation(s)
- Alzbeta Tuerkova
- Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Barbara Zdrazil
- Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| |
Collapse
|
33
|
Khan IH, Javaid M. Big Data Applications in Medical Field: A Literature Review. JOURNAL OF INDUSTRIAL INTEGRATION AND MANAGEMENT 2020. [DOI: 10.1142/s242486222030001x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Digital imaging and medical reporting have acquired an essential role in healthcare, but the main challenge is the storage of a high volume of patient data. Although newer technologies are already introduced in the medical sciences to save records size, Big Data provides advancements by storing a large amount of data to improve the efficiency and quality of patient treatment with better care. It provides intelligent automation capabilities to reduce errors than manual inputs. Large numbers of research papers on big data in the medical field are studied and analyzed for their impacts, benefits, and applications. Big data has great potential to support the digitalization of all medical and clinical records and then save the entire data regarding the medical history of an individual or a group. This paper discusses big data usage for various industries and sectors. Finally, 12 significant applications for the medical field by the implementation of big data are identified and studied with a brief description. This technology can be gainfully used to extract useful information from the available data by analyzing and managing them through a combination of hardware and software. With technological advancement, big data provides health-related information for millions of patient-related to life issues such as lab tests reporting, clinical narratives, demographics, prescription, medical diagnosis, and related documentation. Thus, Big Data is essential in developing a better yet efficient analysis and storage healthcare services. The demand for big data applications is increasing due to its capability of handling and analyzing massive data. Not only in the future but even now, Big Data is proving itself as an axiom of storing, developing, analyzing, and providing overall health information to the physicians.
Collapse
Affiliation(s)
- Ibrahim Haleem Khan
- School of Engineering Sciences and Technology, Jamia Hamdard, New Delhi, India
| | - Mohd Javaid
- Department of Mechanical Engineering, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
34
|
A System Biology-Based Approach for Designing Combination Therapy in Cancer Precision Medicine. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5072697. [PMID: 32908895 PMCID: PMC7471815 DOI: 10.1155/2020/5072697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/07/2020] [Accepted: 07/22/2020] [Indexed: 12/02/2022]
Abstract
In this paper, we have used an agent-based stochastic tumor growth model and presented a mathematical and theoretical perspective to cancer therapy. This perspective can be used to theoretical study of precision medicine and combination therapy in individuals. We have conducted a series of in silico combination therapy experiments. Based on cancer drugs and new findings of cancer biology, we hypothesize relationships between model parameters which in some cases represent individual genome characteristics and cancer drugs, i.e., in our approach, therapy players are delegated by biologically reasonable parameters. In silico experiments showed that combined therapies are more effective when players affect tumor via different mechanisms and have different physical dimensions. This research presents for the first time an algorithm as a theoretical viewpoint for the prediction of effectiveness and classification of therapy sets.
Collapse
|
35
|
Masuzaki R, Kanda T, Sasaki R, Matsumoto N, Nirei K, Ogawa M, Moriyama M. Application of artificial intelligence in hepatology: Minireview. Artif Intell Gastroenterol 2020; 1:5-11. [DOI: 10.35712/aig.v1.i1.5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/23/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023] Open
|
36
|
Bazaga A, Leggate D, Weisser H. Genome-wide investigation of gene-cancer associations for the prediction of novel therapeutic targets in oncology. Sci Rep 2020; 10:10787. [PMID: 32612205 PMCID: PMC7330039 DOI: 10.1038/s41598-020-67846-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/08/2020] [Indexed: 02/03/2023] Open
Abstract
A major cause of failed drug discovery programs is suboptimal target selection, resulting in the development of drug candidates that are potent inhibitors, but ineffective at treating the disease. In the genomics era, the availability of large biomedical datasets with genome-wide readouts has the potential to transform target selection and validation. In this study we investigate how computational intelligence methods can be applied to predict novel therapeutic targets in oncology.
We compared different machine learning classifiers applied to the task of drug target classification for nine different human cancer types. For each cancer type, a set of “known” target genes was obtained and equally-sized sets of “non-targets” were sampled multiple times from the human protein-coding genes. Models were trained on mutation, gene expression (TCGA), and gene essentiality (DepMap) data. In addition, we generated a numerical embedding of the interaction network of protein-coding genes using deep network representation learning and included the results in the modeling. We assessed feature importance using a random forests classifier and performed feature selection based on measuring permutation importance against a null distribution. Our best models achieved good generalization performance based on the AUROC metric. With the best model for each cancer type, we ran predictions on more than 15,000 protein-coding genes to identify potential novel targets. Our results indicate that this approach may be useful to inform early stages of the drug discovery pipeline.
Collapse
Affiliation(s)
- Adrián Bazaga
- Department of Genetics, University of Cambridge, Cambridge, UK. .,STORM Therapeutics Ltd, Cambridge, UK.
| | | | | |
Collapse
|
37
|
Hokken TW, Ribeiro JM, De Jaegere PP, Van Mieghem NM. Precision Medicine in Interventional Cardiology. ACTA ACUST UNITED AC 2020; 15:e03. [PMID: 32382319 PMCID: PMC7203877 DOI: 10.15420/icr.2019.23] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/31/2020] [Indexed: 12/17/2022]
Abstract
Precision medicine has recently become widely advocated. It revolves around the individual patient, taking into account genetic, biomarker, phenotypic or psychosocial characteristics and uses biological, mechanical and/or personal variables to optimise individual therapy. In silico testing, such as the Virtual Physiological Human project, is being promoted to predict risk and to test treatments and medical devices. It combines artificial intelligence and computational modelling to select the best therapeutic option for the individual patient.
Collapse
Affiliation(s)
- Thijmen W Hokken
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Joana M Ribeiro
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center Rotterdam, the Netherlands.,Department of Cardiology, Centro Hospitlar and Universitário de Coimbra Coimbra, Portugal
| | - Peter P De Jaegere
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Nicolas M Van Mieghem
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center Rotterdam, the Netherlands
| |
Collapse
|
38
|
Sánchez López JD, Cambil Martín J, Villegas Calvo M, Luque Martínez F. [Artificial intelligence in healthcare. Are the patient rights protected?]. J Healthc Qual Res 2020; 36:378-379. [PMID: 32179018 DOI: 10.1016/j.jhqr.2019.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 07/30/2019] [Indexed: 11/24/2022]
Affiliation(s)
- J D Sánchez López
- Servicio de Cirugía Oral y Maxilofacial, Centro de Rehabilitación y Traumatología, Hospital Universitario Virgen de las Nieves, Granada, España; Comité Ético de Investigación de Granada, Granada, España.
| | - J Cambil Martín
- Departamento de Enfermería, Facultad de Ciencias de la Salud, Universidad de Granada, Granada, España
| | | | - F Luque Martínez
- Comité Ético de Investigación de Granada, Granada, España; Hospital Universitario Virgen de las Nieves, Granada, España
| |
Collapse
|
39
|
Shahin MH, Bhattacharya S, Silva D, Kim S, Burton J, Podichetty J, Romero K, Conrado DJ. Open Data Revolution in Clinical Research: Opportunities and Challenges. Clin Transl Sci 2020; 13:665-674. [PMID: 32004409 PMCID: PMC7359943 DOI: 10.1111/cts.12756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/05/2019] [Indexed: 01/24/2023] Open
Abstract
Efforts for sharing individual clinical data are gaining momentum due to a heightened recognition that integrated data sets can catalyze biomedical discoveries and drug development. Among the benefits are the fact that data sharing can help generate and investigate new research hypothesis beyond those explored in the original study. Despite several accomplishments establishing public systems and guidance for data sharing in clinical trials, this practice is not the norm. Among the reasons are ethical challenges, such as privacy of individuals, data ownership, and control. This paper creates awareness of the potential benefits and challenges of sharing individual clinical data, how to overcome these challenges, and how as a clinical pharmacology community we can shape future directions in this field.
Collapse
Affiliation(s)
| | - Sanchita Bhattacharya
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California, USA.,Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Diego Silva
- Faculty of Health Sciences, Simon Fraser University, Vancouver, British Columbia, Canada.,Sydney Health Ethics, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Sarah Kim
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | | | | | | | |
Collapse
|