1
|
Mukherjee A, Abraham S, Singh A, Balaji S, Mukunthan KS. From Data to Cure: A Comprehensive Exploration of Multi-omics Data Analysis for Targeted Therapies. Mol Biotechnol 2025; 67:1269-1289. [PMID: 38565775 PMCID: PMC11928429 DOI: 10.1007/s12033-024-01133-6] [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: 12/27/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
In the dynamic landscape of targeted therapeutics, drug discovery has pivoted towards understanding underlying disease mechanisms, placing a strong emphasis on molecular perturbations and target identification. This paradigm shift, crucial for drug discovery, is underpinned by big data, a transformative force in the current era. Omics data, characterized by its heterogeneity and enormity, has ushered biological and biomedical research into the big data domain. Acknowledging the significance of integrating diverse omics data strata, known as multi-omics studies, researchers delve into the intricate interrelationships among various omics layers. This review navigates the expansive omics landscape, showcasing tailored assays for each molecular layer through genomes to metabolomes. The sheer volume of data generated necessitates sophisticated informatics techniques, with machine-learning (ML) algorithms emerging as robust tools. These datasets not only refine disease classification but also enhance diagnostics and foster the development of targeted therapeutic strategies. Through the integration of high-throughput data, the review focuses on targeting and modeling multiple disease-regulated networks, validating interactions with multiple targets, and enhancing therapeutic potential using network pharmacology approaches. Ultimately, this exploration aims to illuminate the transformative impact of multi-omics in the big data era, shaping the future of biological research.
Collapse
Affiliation(s)
- Arnab Mukherjee
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| | - Suzanna Abraham
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| | - Akshita Singh
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| | - S Balaji
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| | - K S Mukunthan
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
2
|
Spooner A, Moridani MK, Toplis B, Behary J, Safarchi A, Maher S, Vafaee F, Zekry A, Sowmya A. Benchmarking ensemble machine learning algorithms for multi-class, multi-omics data integration in clinical outcome prediction. Brief Bioinform 2025; 26:bbaf116. [PMID: 40116658 PMCID: PMC11926982 DOI: 10.1093/bib/bbaf116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/06/2025] [Accepted: 02/21/2025] [Indexed: 03/23/2025] Open
Abstract
The complementary information found in different modalities of patient data can aid in more accurate modelling of a patient's disease state and a better understanding of the underlying biological processes of a disease. However, the analysis of multi-modal, multi-omics data presents many challenges. In this work, we compare the performance of a variety of ensemble machine learning (ML) algorithms that are capable of late integration of multi-class data from different modalities. The ensemble methods and their variations tested were (i) a voting ensemble, with hard and soft vote, (ii) a meta learner, and (iii) a multi-modal AdaBoost model using hard vote, soft vote, and meta learner to integrate the modalities on each boosting round, the PB-MVBoost model and a novel application of a mixture of expert's model. These were compared to simple concatenation. We examine these methods using data from an in-house study on hepatocellular carcinoma, plus validation datasets on studies from breast cancer and irritable bowel disease. We develop models that achieve an area under the receiver operating curve of up to 0.85 and find that two boosted methods, PB-MVBoost and AdaBoost with soft vote were the best performing models. We also examine the stability of features selected and the size of the clinical signature. Our work shows that integrating complementary omics and data modalities with effective ensemble ML models enhances accuracy in multi-class clinical outcome predictions and produces more stable predictive features than individual modalities or simple concatenation. We provide recommendations for the integration of multi-modal multi-class data.
Collapse
Affiliation(s)
- Annette Spooner
- School of Computer Science and Engineering, University of New South Wales, High St, Kensington, NSW 2052, Australia
| | - Mohammad Karimi Moridani
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, NSW 2052, Australia
| | - Barbra Toplis
- St George and Sutherland Clinical Campuses, University of New South Wales, Short St, Kogarah, NSW 2217, Australia
| | - Jason Behary
- St George and Sutherland Clinical Campuses, University of New South Wales, Short St, Kogarah, NSW 2217, Australia
- Department of Gastroenterology and Hepatology, St George Hospital, Gray St, Kogarah, NSW 2217, Australia
| | - Azadeh Safarchi
- Health and Biosecurity, Microbiome for One System Health, Commonwealth Scientific and Industrial Research Organisation, 160 Hawkesbury Rd, Westmead, NSW 2145, Australia
| | - Salim Maher
- St George and Sutherland Clinical Campuses, University of New South Wales, Short St, Kogarah, NSW 2217, Australia
- Department of Gastroenterology and Hepatology, St George Hospital, Gray St, Kogarah, NSW 2217, Australia
| | - Fatemeh Vafaee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, NSW 2052, Australia
- UNSW Data Science Hub, University of New South Wales, High St, Kensington, NSW 2052, Australia
| | - Amany Zekry
- St George and Sutherland Clinical Campuses, University of New South Wales, Short St, Kogarah, NSW 2217, Australia
- Department of Gastroenterology and Hepatology, St George Hospital, Gray St, Kogarah, NSW 2217, Australia
| | - Arcot Sowmya
- School of Computer Science and Engineering, University of New South Wales, High St, Kensington, NSW 2052, Australia
| |
Collapse
|
3
|
Chicco D, Fabris A, Jurman G. The Venus score for the assessment of the quality and trustworthiness of biomedical datasets. BioData Min 2025; 18:1. [PMID: 39780220 PMCID: PMC11716409 DOI: 10.1186/s13040-024-00412-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Biomedical datasets are the mainstays of computational biology and health informatics projects, and can be found on multiple data platforms online or obtained from wet-lab biologists and physicians. The quality and the trustworthiness of these datasets, however, can sometimes be poor, producing bad results in turn, which can harm patients and data subjects. To address this problem, policy-makers, researchers, and consortia have proposed diverse regulations, guidelines, and scores to assess the quality and increase the reliability of datasets. Although generally useful, however, they are often incomplete and impractical. The guidelines of Datasheets for Datasets, in particular, are too numerous; the requirements of the Kaggle Dataset Usability Score focus on non-scientific requisites (for example, including a cover image); and the European Union Artificial Intelligence Act (EU AI Act) sets forth sparse and general data governance requirements, which we tailored to datasets for biomedical AI. Against this backdrop, we introduce our new Venus score to assess the data quality and trustworthiness of biomedical datasets. Our score ranges from 0 to 10 and consists of ten questions that anyone developing a bioinformatics, medical informatics, or cheminformatics dataset should answer before the release. In this study, we first describe the EU AI Act, Datasheets for Datasets, and the Kaggle Dataset Usability Score, presenting their requirements and their drawbacks. To do so, we reverse-engineer the weights of the influential Kaggle Score for the first time and report them in this study. We distill the most important data governance requirements into ten questions tailored to the biomedical domain, comprising the Venus score. We apply the Venus score to twelve datasets from multiple subdomains, including electronic health records, medical imaging, microarray and bulk RNA-seq gene expression, cheminformatics, physiologic electrogram signals, and medical text. Analyzing the results, we surface fine-grained strengths and weaknesses of popular datasets, as well as aggregate trends. Most notably, we find a widespread tendency to gloss over sources of data inaccuracy and noise, which may hinder the reliable exploitation of data and, consequently, research results. Overall, our results confirm the applicability and utility of the Venus score to assess the trustworthiness of biomedical data.
Collapse
Affiliation(s)
- Davide Chicco
- Università di Milano-Bicocca & University of Toronto, Toronto, Canada.
| | | | | |
Collapse
|
4
|
Briscik M, Tazza G, Vidács L, Dillies MA, Déjean S. Supervised multiple kernel learning approaches for multi-omics data integration. BioData Min 2024; 17:53. [PMID: 39580456 PMCID: PMC11585117 DOI: 10.1186/s13040-024-00406-9] [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: 03/22/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Advances in high-throughput technologies have originated an ever-increasing availability of omics datasets. The integration of multiple heterogeneous data sources is currently an issue for biology and bioinformatics. Multiple kernel learning (MKL) has shown to be a flexible and valid approach to consider the diverse nature of multi-omics inputs, despite being an underused tool in genomic data mining. RESULTS We provide novel MKL approaches based on different kernel fusion strategies. To learn from the meta-kernel of input kernels, we adapted unsupervised integration algorithms for supervised tasks with support vector machines. We also tested deep learning architectures for kernel fusion and classification. The results show that MKL-based models can outperform more complex, state-of-the-art, supervised multi-omics integrative approaches. CONCLUSION Multiple kernel learning offers a natural framework for predictive models in multi-omics data. It proved to provide a fast and reliable solution that can compete with and outperform more complex architectures. Our results offer a direction for bio-data mining research, biomarker discovery and further development of methods for heterogeneous data integration.
Collapse
Affiliation(s)
- Mitja Briscik
- Institut de Mathématiques de Toulouse, UMR5219, CNRS, UPS, Université de Toulouse, Cedex 9, Toulouse, 31062, France.
| | - Gabriele Tazza
- Department of Computer Science, Applied Artificial Intelligence Group , University of Szeged, Szeged, 6720, Hungary.
| | - László Vidács
- Department of Computer Science, Applied Artificial Intelligence Group , University of Szeged, Szeged, 6720, Hungary
| | - Marie-Agnès Dillies
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015, Paris, France
| | - Sébastien Déjean
- Institut de Mathématiques de Toulouse, UMR5219, CNRS, UPS, Université de Toulouse, Cedex 9, Toulouse, 31062, France
| |
Collapse
|
5
|
Son A, Park J, Kim W, Yoon Y, Lee S, Ji J, Kim H. Recent Advances in Omics, Computational Models, and Advanced Screening Methods for Drug Safety and Efficacy. TOXICS 2024; 12:822. [PMID: 39591001 PMCID: PMC11598288 DOI: 10.3390/toxics12110822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/10/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024]
Abstract
It is imperative to comprehend the mechanisms that underlie drug toxicity in order to enhance the efficacy and safety of novel therapeutic agents. The capacity to identify molecular pathways that contribute to drug-induced toxicity has been significantly enhanced by recent developments in omics technologies, such as transcriptomics, proteomics, and metabolomics. This has enabled the early identification of potential adverse effects. These insights are further enhanced by computational tools, including quantitative structure-activity relationship (QSAR) analyses and machine learning models, which accurately predict toxicity endpoints. Additionally, technologies such as physiologically based pharmacokinetic (PBPK) modeling and micro-physiological systems (MPS) provide more precise preclinical-to-clinical translation, thereby improving drug safety assessments. This review emphasizes the synergy between sophisticated screening technologies, in silico modeling, and omics data, emphasizing their roles in reducing late-stage drug development failures. Challenges persist in the integration of a variety of data types and the interpretation of intricate biological interactions, despite the progress that has been made. The development of standardized methodologies that further enhance predictive toxicology is contingent upon the ongoing collaboration between researchers, clinicians, and regulatory bodies. This collaboration ensures the development of therapeutic pharmaceuticals that are more effective and safer.
Collapse
Affiliation(s)
- Ahrum Son
- Department of Molecular Medicine, Scripps Research, San Diego, CA 92037, USA;
| | - Jongham Park
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (Y.Y.); (S.L.)
| | - Woojin Kim
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (Y.Y.); (S.L.)
| | - Yoonki Yoon
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (Y.Y.); (S.L.)
| | - Sangwoon Lee
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (Y.Y.); (S.L.)
| | - Jaeho Ji
- Department of Convergent Bioscience and Informatics, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea;
| | - Hyunsoo Kim
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (Y.Y.); (S.L.)
- Department of Convergent Bioscience and Informatics, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea;
- Protein AI Design Institute, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
- SCICS, Prove Beyond AI, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| |
Collapse
|
6
|
Ortega-Vallbona R, Palomino-Schätzlein M, Tolosa L, Benfenati E, Ecker GF, Gozalbes R, Serrano-Candelas E. Computational Strategies for Assessing Adverse Outcome Pathways: Hepatic Steatosis as a Case Study. Int J Mol Sci 2024; 25:11154. [PMID: 39456937 PMCID: PMC11508863 DOI: 10.3390/ijms252011154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The evolving landscape of chemical risk assessment is increasingly focused on developing tiered, mechanistically driven approaches that avoid the use of animal experiments. In this context, adverse outcome pathways have gained importance for evaluating various types of chemical-induced toxicity. Using hepatic steatosis as a case study, this review explores the use of diverse computational techniques, such as structure-activity relationship models, quantitative structure-activity relationship models, read-across methods, omics data analysis, and structure-based approaches to fill data gaps within adverse outcome pathway networks. Emphasizing the regulatory acceptance of each technique, we examine how these methodologies can be integrated to provide a comprehensive understanding of chemical toxicity. This review highlights the transformative impact of in silico techniques in toxicology, proposing guidelines for their application in evidence gathering for developing and filling data gaps in adverse outcome pathway networks. These guidelines can be applied to other cases, advancing the field of toxicological risk assessment.
Collapse
Affiliation(s)
- Rita Ortega-Vallbona
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
| | - Martina Palomino-Schätzlein
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
| | - Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, 46026 Valencia, Spain;
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, C/Monforte de Lemos, 28029 Madrid, Spain
| | - Emilio Benfenati
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
| | - Gerhard F. Ecker
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek Platz 2, 1090 Wien, Austria;
| | - Rafael Gozalbes
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
- MolDrug AI Systems S.L., Olimpia Arozena Torres 45, 46108 Valencia, Spain
| | - Eva Serrano-Candelas
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
| |
Collapse
|
7
|
Liu W, He H, Chicco D. Gene signatures for cancer research: A 25-year retrospective and future avenues. PLoS Comput Biol 2024; 20:e1012512. [PMID: 39413055 PMCID: PMC11482671 DOI: 10.1371/journal.pcbi.1012512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024] Open
Abstract
Over the past two decades, extensive studies, particularly in cancer analysis through large datasets like The Cancer Genome Atlas (TCGA), have aimed at improving patient therapies and precision medicine. However, limited overlap and inconsistencies among gene signatures across different cohorts pose challenges. The dynamic nature of the transcriptome, encompassing diverse RNA species and functional complexities at gene and isoform levels, introduces intricacies, and current gene signatures face reproducibility issues due to the unique transcriptomic landscape of each patient. In this context, discrepancies arising from diverse sequencing technologies, data analysis algorithms, and software tools further hinder consistency. While careful experimental design, analytical strategies, and standardized protocols could enhance reproducibility, future prospects lie in multiomics data integration, machine learning techniques, open science practices, and collaborative efforts. Standardized metrics, quality control measures, and advancements in single-cell RNA-seq will contribute to unbiased gene signature identification. In this perspective article, we outline some thoughts and insights addressing challenges, standardized practices, and advanced methodologies enhancing the reliability of gene signatures in disease transcriptomic research.
Collapse
Affiliation(s)
- Wei Liu
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Huaqin He
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Davide Chicco
- Dipartimento di Informatica Sistemistica e Comunicazione, Università di Milano-Bicocca, Milan, Italy
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Verma S, Magazzù G, Eftekhari N, Lou T, Gilhespy A, Occhipinti A, Angione C. Cross-attention enables deep learning on limited omics-imaging-clinical data of 130 lung cancer patients. CELL REPORTS METHODS 2024; 4:100817. [PMID: 38981473 PMCID: PMC11294841 DOI: 10.1016/j.crmeth.2024.100817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 04/18/2024] [Accepted: 06/17/2024] [Indexed: 07/11/2024]
Abstract
Deep-learning tools that extract prognostic factors derived from multi-omics data have recently contributed to individualized predictions of survival outcomes. However, the limited size of integrated omics-imaging-clinical datasets poses challenges. Here, we propose two biologically interpretable and robust deep-learning architectures for survival prediction of non-small cell lung cancer (NSCLC) patients, learning simultaneously from computed tomography (CT) scan images, gene expression data, and clinical information. The proposed models integrate patient-specific clinical, transcriptomic, and imaging data and incorporate Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome pathway information, adding biological knowledge within the learning process to extract prognostic gene biomarkers and molecular pathways. While both models accurately stratify patients in high- and low-risk groups when trained on a dataset of only 130 patients, introducing a cross-attention mechanism in a sparse autoencoder significantly improves the performance, highlighting tumor regions and NSCLC-related genes as potential biomarkers and thus offering a significant methodological advancement when learning from small imaging-omics-clinical samples.
Collapse
Affiliation(s)
- Suraj Verma
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK
| | | | | | - Thai Lou
- Gateshead Health NHS Foundation Trust, Gateshead, UK
| | - Alex Gilhespy
- South Tyneside and Sunderland NHS Foundation Trust, Sunderland, UK
| | - Annalisa Occhipinti
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK; Centre for Digital Innovation, Teesside University, Middlesbrough, UK; National Horizons Centre, Teesside University, Darlington, UK
| | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK; Centre for Digital Innovation, Teesside University, Middlesbrough, UK; National Horizons Centre, Teesside University, Darlington, UK.
| |
Collapse
|
9
|
Valous NA, Popp F, Zörnig I, Jäger D, Charoentong P. Graph machine learning for integrated multi-omics analysis. Br J Cancer 2024; 131:205-211. [PMID: 38729996 PMCID: PMC11263675 DOI: 10.1038/s41416-024-02706-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
Multi-omics experiments at bulk or single-cell resolution facilitate the discovery of hypothesis-generating biomarkers for predicting response to therapy, as well as aid in uncovering mechanistic insights into cellular and microenvironmental processes. Many methods for data integration have been developed for the identification of key elements that explain or predict disease risk or other biological outcomes. The heterogeneous graph representation of multi-omics data provides an advantage for discerning patterns suitable for predictive/exploratory analysis, thus permitting the modeling of complex relationships. Graph-based approaches-including graph neural networks-potentially offer a reliable methodological toolset that can provide a tangible alternative to scientists and clinicians that seek ideas and implementation strategies in the integrated analysis of their omics sets for biomedical research. Graph-based workflows continue to push the limits of the technological envelope, and this perspective provides a focused literature review of research articles in which graph machine learning is utilized for integrated multi-omics data analyses, with several examples that demonstrate the effectiveness of graph-based approaches.
Collapse
Affiliation(s)
- Nektarios A Valous
- Applied Tumor Immunity Clinical Cooperation Unit, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany.
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany.
| | - Ferdinand Popp
- Applied Tumor Immunity Clinical Cooperation Unit, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Inka Zörnig
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Dirk Jäger
- Applied Tumor Immunity Clinical Cooperation Unit, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Pornpimol Charoentong
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| |
Collapse
|
10
|
Shannon CP, Lee AH, Tebbutt SJ, Singh A. A Commentary on Multi-omics Data Integration in Systems Vaccinology. J Mol Biol 2024; 436:168522. [PMID: 38458605 DOI: 10.1016/j.jmb.2024.168522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Affiliation(s)
| | - Amy Hy Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Scott J Tebbutt
- PROOF Centre of Excellence, Vancouver, Canada; Department of Medicine, The University of British Columbia, Vancouver, Canada; Centre for Heart Lung Innovation, Vancouver, Canada
| | - Amrit Singh
- Centre for Heart Lung Innovation, Vancouver, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, Canada.
| |
Collapse
|
11
|
Chicco D, Spolaor S, Nobile MS. Ten quick tips for fuzzy logic modeling of biomedical systems. PLoS Comput Biol 2023; 19:e1011700. [PMID: 38127800 PMCID: PMC10734980 DOI: 10.1371/journal.pcbi.1011700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Fuzzy logic is useful tool to describe and represent biological or medical scenarios, where often states and outcomes are not only completely true or completely false, but rather partially true or partially false. Despite its usefulness and spread, fuzzy logic modeling might easily be done in the wrong way, especially by beginners and unexperienced researchers, who might overlook some important aspects or might make common mistakes. Malpractices and pitfalls, in turn, can lead to wrong or overoptimistic, inflated results, with negative consequences to the biomedical research community trying to comprehend a particular phenomenon, or even to patients suffering from the investigated disease. To avoid common mistakes, we present here a list of quick tips for fuzzy logic modeling any biomedical scenario: some guidelines which should be taken into account by any fuzzy logic practitioner, including experts. We believe our best practices can have a strong impact in the scientific community, allowing researchers who follow them to obtain better, more reliable results and outcomes in biomedical contexts.
Collapse
Affiliation(s)
- Davide Chicco
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Dipartimento di Informatica Sistemistica e Comunicazione, Università di Milano-Bicocca, Milan, Italy
| | - Simone Spolaor
- Microsystems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Marco S. Nobile
- Department of Environmental Sciences Informatics and Statistics, Ca’ Foscari University of Venice, Venice, Italy
| |
Collapse
|