ijms-logo

Journal Browser

Journal Browser

Artificial Intelligence in Biomarker Discovery

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Informatics".

Deadline for manuscript submissions: closed (15 January 2023) | Viewed by 21140

Special Issue Editor


E-Mail Website
Guest Editor
Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece
Interests: biomarker; classifier; liquid biopsy; epigenetics; cell culture; solid tumor; diagnosis; moniroting; neuropeptide; drug target; drug response; metabolic disease; diabetes
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues, 

Biomarkers are the cornerstone of precision medicine: identified as measurable indicators of some biological state or condition, they promise to offer solutions for accurate diagnosis, prognosis, and therapeutic monitoring. Over the last 2 decades, the high throughput technology in biomedicine has demonstrated significant advancements, leading to a vast accumulation of publicly available precious x-omics high dimension biological datasets, presenting welcoming opportunities for new science. Innovative bioinformatics Artificial Intelligence (AI) tools promise to enable exploitation of these data, shifting the global scientific trend from hypothesis to data-driven approaches. Modern statistical and machine learning methods perform predictive modeling and knowledge discovery. This brings unprecedented added value in different medical conditions, gaining intuition into the molecular pathological mechanisms of disease, identifying novel drug targets, or designing emerging cost-effective assays for improving diagnosis, prognosis, or response prediction, readily available to clinical application. This Special Issue plans to offer an overview of recent advances in data-driven approaches aided state-of-the-art AI in diverse areas of biomedical research, with emphasis on biomarker discovery.

Potential topics include but are not limited to machine learning for biomarkers in precision oncology, machine learning tools for feature selection or dimensionality reduction in molecular biology, text mining tools and pathway analysis for biomarker exploration, artificial intelligence in microbiome analysis, artificial intelligence in epigenetic biomarkers and epitranscriptomics, machine learning tools in drug discovery, biomarker retrieval and knowledge reasoning systems in molecular biology, and data integration in biomedical or biomarker research.

Prof. Dr. Ekaterini Chatzaki
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • machine learning
  • biomarker
  • precision medicine
  • artificial intelligence
  • text mining
  • automation
  • genomics
  • epigenomics
  • proteomics
  • big data
  • clinical
  • diagnostic
  • bioinformatic

Related Special Issue

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

17 pages, 3370 KiB  
Article
Classification of COVID-19 Patients into Clinically Relevant Subsets by a Novel Machine Learning Pipeline Using Transcriptomic Features
by Andrea R. Daamen, Prathyusha Bachali, Amrie C. Grammer and Peter E. Lipsky
Int. J. Mol. Sci. 2023, 24(5), 4905; https://doi.org/10.3390/ijms24054905 - 03 Mar 2023
Cited by 5 | Viewed by 1714
Abstract
The persistent impact of the COVID-19 pandemic and heterogeneity in disease manifestations point to a need for innovative approaches to identify drivers of immune pathology and predict whether infected patients will present with mild/moderate or severe disease. We have developed a novel iterative [...] Read more.
The persistent impact of the COVID-19 pandemic and heterogeneity in disease manifestations point to a need for innovative approaches to identify drivers of immune pathology and predict whether infected patients will present with mild/moderate or severe disease. We have developed a novel iterative machine learning pipeline that utilizes gene enrichment profiles from blood transcriptome data to stratify COVID-19 patients based on disease severity and differentiate severe COVID cases from other patients with acute hypoxic respiratory failure. The pattern of gene module enrichment in COVID-19 patients overall reflected broad cellular expansion and metabolic dysfunction, whereas increased neutrophils, activated B cells, T-cell lymphopenia, and proinflammatory cytokine production were specific to severe COVID patients. Using this pipeline, we also identified small blood gene signatures indicative of COVID-19 diagnosis and severity that could be used as biomarker panels in the clinical setting. Full article
(This article belongs to the Special Issue Artificial Intelligence in Biomarker Discovery)
Show Figures

Figure 1

12 pages, 2051 KiB  
Article
A Receptor Tyrosine Kinase Inhibitor Sensitivity Prediction Model Identifies AXL Dependency in Leukemia
by Ahmad Nasimian, Lina Al Ashiri, Mehreen Ahmed, Hongzhi Duan, Xiaoyue Zhang, Lars Rönnstrand and Julhash U. Kazi
Int. J. Mol. Sci. 2023, 24(4), 3830; https://doi.org/10.3390/ijms24043830 - 14 Feb 2023
Cited by 3 | Viewed by 1831
Abstract
Despite incredible progress in cancer treatment, therapy resistance remains the leading limiting factor for long-term survival. During drug treatment, several genes are transcriptionally upregulated to mediate drug tolerance. Using highly variable genes and pharmacogenomic data for acute myeloid leukemia (AML), we developed a [...] Read more.
Despite incredible progress in cancer treatment, therapy resistance remains the leading limiting factor for long-term survival. During drug treatment, several genes are transcriptionally upregulated to mediate drug tolerance. Using highly variable genes and pharmacogenomic data for acute myeloid leukemia (AML), we developed a drug sensitivity prediction model for the receptor tyrosine kinase inhibitor sorafenib and achieved more than 80% prediction accuracy. Furthermore, by using Shapley additive explanations for determining leading features, we identified AXL as an important feature for drug resistance. Drug-resistant patient samples displayed enrichment of protein kinase C (PKC) signaling, which was also identified in sorafenib-treated FLT3-ITD-dependent AML cell lines by a peptide-based kinase profiling assay. Finally, we show that pharmacological inhibition of tyrosine kinase activity enhances AXL expression, phosphorylation of the PKC-substrate cyclic AMP response element binding (CREB) protein, and displays synergy with AXL and PKC inhibitors. Collectively, our data suggest an involvement of AXL in tyrosine kinase inhibitor resistance and link PKC activation as a possible signaling mediator. Full article
(This article belongs to the Special Issue Artificial Intelligence in Biomarker Discovery)
Show Figures

Figure 1

14 pages, 10219 KiB  
Article
Machine Learning Quantified Tumor-Stroma Ratio Is an Independent Prognosticator in Muscle-Invasive Bladder Cancer
by Qingyuan Zheng, Zhengyu Jiang, Xinmiao Ni, Song Yang, Panpan Jiao, Jiejun Wu, Lin Xiong, Jingping Yuan, Jingsong Wang, Jun Jian, Lei Wang, Rui Yang, Zhiyuan Chen and Xiuheng Liu
Int. J. Mol. Sci. 2023, 24(3), 2746; https://doi.org/10.3390/ijms24032746 - 01 Feb 2023
Cited by 6 | Viewed by 1993
Abstract
Although the tumor-stroma ratio (TSR) has prognostic value in many cancers, the traditional semi-quantitative visual assessment method has inter-observer variability, making it impossible for clinical practice. We aimed to develop a machine learning (ML) algorithm for accurately quantifying TSR in hematoxylin-and-eosin (H&E)-stained whole [...] Read more.
Although the tumor-stroma ratio (TSR) has prognostic value in many cancers, the traditional semi-quantitative visual assessment method has inter-observer variability, making it impossible for clinical practice. We aimed to develop a machine learning (ML) algorithm for accurately quantifying TSR in hematoxylin-and-eosin (H&E)-stained whole slide images (WSI) and further investigate its prognostic effect in patients with muscle-invasive bladder cancer (MIBC). We used an optimal cell classifier previously built based on QuPath open-source software and ML algorithm for quantitative calculation of TSR. We retrospectively analyzed data from two independent cohorts to verify the prognostic significance of ML-based TSR in MIBC patients. WSIs from 133 MIBC patients were used as the discovery set to identify the optimal association of TSR with patient survival outcomes. Furthermore, we performed validation in an independent external cohort consisting of 261 MIBC patients. We demonstrated a significant prognostic association of ML-based TSR with survival outcomes in MIBC patients (p < 0.001 for all comparisons), with higher TSR associated with better prognosis. Uni- and multivariate Cox regression analyses showed that TSR was independently associated with overall survival (p < 0.001 for all analyses) after adjusting for clinicopathological factors including age, gender, and pathologic stage. TSR was found to be a strong prognostic factor that was not redundant with the existing staging system in different subgroup analyses (p < 0.05 for all analyses). Finally, the expression of six genes (DACH1, DEEND2A, NOTCH4, DTWD1, TAF6L, and MARCHF5) were significantly associated with TSR, revealing possible potential biological relevance. In conclusion, we developed an ML algorithm based on WSIs of MIBC patients to accurately quantify TSR and demonstrated its prognostic validity for MIBC patients in two independent cohorts. This objective quantitative method allows application in clinical practice while reducing the workload of pathologists. Thus, it might be of significant aid in promoting precise pathology services in MIBC. Full article
(This article belongs to the Special Issue Artificial Intelligence in Biomarker Discovery)
Show Figures

Figure 1

8 pages, 1238 KiB  
Article
Prediction and Ranking of Biomarkers Using multiple UniReD
by Ismini Baltsavia, Theodosios Theodosiou, Nikolas Papanikolaou, Georgios A. Pavlopoulos, Grigorios D. Amoutzias, Maria Panagopoulou, Ekaterini Chatzaki, Evangelos Andreakos and Ioannis Iliopoulos
Int. J. Mol. Sci. 2022, 23(19), 11112; https://doi.org/10.3390/ijms231911112 - 21 Sep 2022
Viewed by 1256
Abstract
Protein–protein interactions (PPIs) are of key importance for understanding how cells and organisms function. Thus, in recent decades, many approaches have been developed for the identification and discovery of such interactions. These approaches addressed the problem of PPI identification either by an experimental [...] Read more.
Protein–protein interactions (PPIs) are of key importance for understanding how cells and organisms function. Thus, in recent decades, many approaches have been developed for the identification and discovery of such interactions. These approaches addressed the problem of PPI identification either by an experimental point of view or by a computational one. Here, we present an updated version of UniReD, a computational prediction tool which takes advantage of biomedical literature aiming to extract documented, already published protein associations and predict undocumented ones. The usefulness of this computational tool has been previously evaluated by experimentally validating predicted interactions and by benchmarking it against public databases of experimentally validated PPIs. In its updated form, UniReD allows the user to provide a list of proteins of known implication in, e.g., a particular disease, as well as another list of proteins that are potentially associated with the proteins of the first list. UniReD then automatically analyzes both lists and ranks the proteins of the second list by their association with the proteins of the first list, thus serving as a potential biomarker discovery/validation tool. Full article
(This article belongs to the Special Issue Artificial Intelligence in Biomarker Discovery)
Show Figures

Figure 1

25 pages, 4350 KiB  
Article
Outcome Prediction in Critically-Ill Patients with Venous Thromboembolism and/or Cancer Using Machine Learning Algorithms: External Validation and Comparison with Scoring Systems
by Vasiliki Danilatou, Stylianos Nikolakakis, Despoina Antonakaki, Christos Tzagkarakis, Dimitrios Mavroidis, Theodoros Kostoulas and Sotirios Ioannidis
Int. J. Mol. Sci. 2022, 23(13), 7132; https://doi.org/10.3390/ijms23137132 - 27 Jun 2022
Cited by 7 | Viewed by 2764
Abstract
Intensive care unit (ICU) patients with venous thromboembolism (VTE) and/or cancer suffer from high mortality rates. Mortality prediction in the ICU has been a major medical challenge for which several scoring systems exist but lack in specificity. This study focuses on two target [...] Read more.
Intensive care unit (ICU) patients with venous thromboembolism (VTE) and/or cancer suffer from high mortality rates. Mortality prediction in the ICU has been a major medical challenge for which several scoring systems exist but lack in specificity. This study focuses on two target groups, namely patients with thrombosis or cancer. The main goal is to develop and validate interpretable machine learning (ML) models to predict early and late mortality, while exploiting all available data stored in the medical record. To this end, retrospective data from two freely accessible databases, MIMIC-III and eICU, were used. Well-established ML algorithms were implemented utilizing automated and purposely built ML frameworks for addressing class imbalance. Prediction of early mortality showed excellent performance in both disease categories, in terms of the area under the receiver operating characteristic curve (AUCROC): VTE-MIMIC-III 0.93, eICU 0.87, cancer-MIMIC-III 0.94. On the other hand, late mortality prediction showed lower performance, i.e., AUCROC: VTE 0.82, cancer 0.740.88. The predictive model of early mortality developed from 1651 VTE patients (MIMIC-III) ended up with a signature of 35 features and was externally validated in 2659 patients from the eICU dataset. Our model outperformed traditional scoring systems in predicting early as well as late mortality. Novel biomarkers, such as red cell distribution width, were identified.
Full article
(This article belongs to the Special Issue Artificial Intelligence in Biomarker Discovery)
Show Figures

Figure 1

22 pages, 22717 KiB  
Article
Tissue-Specific Methylation Biosignatures for Monitoring Diseases: An In Silico Approach
by Makrina Karaglani, Maria Panagopoulou, Ismini Baltsavia, Paraskevi Apalaki, Theodosis Theodosiou, Ioannis Iliopoulos, Ioannis Tsamardinos and Ekaterini Chatzaki
Int. J. Mol. Sci. 2022, 23(6), 2959; https://doi.org/10.3390/ijms23062959 - 09 Mar 2022
Cited by 5 | Viewed by 2407
Abstract
Tissue-specific gene methylation events are key to the pathogenesis of several diseases and can be utilized for diagnosis and monitoring. Here, we established an in silico pipeline to analyze high-throughput methylome datasets to identify specific methylation fingerprints in three pathological entities of major [...] Read more.
Tissue-specific gene methylation events are key to the pathogenesis of several diseases and can be utilized for diagnosis and monitoring. Here, we established an in silico pipeline to analyze high-throughput methylome datasets to identify specific methylation fingerprints in three pathological entities of major burden, i.e., breast cancer (BrCa), osteoarthritis (OA) and diabetes mellitus (DM). Differential methylation analysis was conducted to compare tissues/cells related to the pathology and different types of healthy tissues, revealing Differentially Methylated Genes (DMGs). Highly performing and low feature number biosignatures were built with automated machine learning, including: (1) a five-gene biosignature discriminating BrCa tissue from healthy tissues (AUC 0.987 and precision 0.987), (2) three equivalent OA cartilage-specific biosignatures containing four genes each (AUC 0.978 and precision 0.986) and (3) a four-gene pancreatic β-cell-specific biosignature (AUC 0.984 and precision 0.995). Next, the BrCa biosignature was validated using an independent ccfDNA dataset showing an AUC and precision of 1.000, verifying the biosignature’s applicability in liquid biopsy. Functional and protein interaction prediction analysis revealed that most DMGs identified are involved in pathways known to be related to the studied diseases or pointed to new ones. Overall, our data-driven approach contributes to the maximum exploitation of high-throughput methylome readings, helping to establish specific disease profiles to be applied in clinical practice and to understand human pathology. Full article
(This article belongs to the Special Issue Artificial Intelligence in Biomarker Discovery)
Show Figures

Figure 1

Review

Jump to: Research

11 pages, 3106 KiB  
Review
OMOP CDM Can Facilitate Data-Driven Studies for Cancer Prediction: A Systematic Review
by Najia Ahmadi, Yuan Peng, Markus Wolfien, Michéle Zoch and Martin Sedlmayr
Int. J. Mol. Sci. 2022, 23(19), 11834; https://doi.org/10.3390/ijms231911834 - 05 Oct 2022
Cited by 15 | Viewed by 3691
Abstract
The current generation of sequencing technologies has led to significant advances in identifying novel disease-associated mutations and generated large amounts of data in a high-throughput manner. Such data in conjunction with clinical routine data are proven to be highly useful in deriving population-level [...] Read more.
The current generation of sequencing technologies has led to significant advances in identifying novel disease-associated mutations and generated large amounts of data in a high-throughput manner. Such data in conjunction with clinical routine data are proven to be highly useful in deriving population-level and patient-level predictions, especially in the field of cancer precision medicine. However, data harmonization across multiple national and international clinical sites is an essential step for the assessment of events and outcomes associated with patients, which is currently not adequately addressed. The Observational Medical Outcomes Partnership (OMOP) Common Data Model (CDM) is an internationally established research data repository introduced by the Observational Health Data Science and Informatics (OHDSI) community to overcome this issue. To address the needs of cancer research, the genomic vocabulary extension was introduced in 2020 to support the standardization of subsequent data analysis. In this review, we evaluate the current potential of the OMOP CDM to be applicable in cancer prediction and how comprehensively the genomic vocabulary extension of the OMOP can serve current needs of AI-based predictions. For this, we systematically screened the literature for articles that use the OMOP CDM in predictive analyses in cancer and investigated the underlying predictive models/tools. Interestingly, we found 248 articles, of which most use the OMOP for harmonizing their data, but only 5 make use of predictive algorithms on OMOP-based data and fulfill our criteria. The studies present multicentric investigations, in which the OMOP played an essential role in discovering and optimizing machine learning (ML)-based models. Ultimately, the use of the OMOP CDM leads to standardized data-driven studies for multiple clinical sites and enables a more solid basis utilizing, e.g., ML models that can be reused and combined in early prediction, diagnosis, and improvement of personalized cancer care and biomarker discovery. Full article
(This article belongs to the Special Issue Artificial Intelligence in Biomarker Discovery)
Show Figures

Figure 1

18 pages, 876 KiB  
Review
Revisiting miRNA Association with Melanoma Recurrence and Metastasis from a Machine Learning Point of View
by Aigli Korfiati, Katerina Grafanaki, George C. Kyriakopoulos, Ilias Skeparnias, Sophia Georgiou, George Sakellaropoulos and Constantinos Stathopoulos
Int. J. Mol. Sci. 2022, 23(3), 1299; https://doi.org/10.3390/ijms23031299 - 24 Jan 2022
Cited by 11 | Viewed by 3282
Abstract
The diagnostic and prognostic value of miRNAs in cutaneous melanoma (CM) has been broadly studied and supported by advanced bioinformatics tools. From early studies using miRNA arrays with several limitations, to the recent NGS-derived miRNA expression profiles, an accurate diagnostic panel of a [...] Read more.
The diagnostic and prognostic value of miRNAs in cutaneous melanoma (CM) has been broadly studied and supported by advanced bioinformatics tools. From early studies using miRNA arrays with several limitations, to the recent NGS-derived miRNA expression profiles, an accurate diagnostic panel of a comprehensive pre-specified set of miRNAs that could aid timely identification of specific cancer stages is still elusive, mainly because of the heterogeneity of the approaches and the samples. Herein, we summarize the existing studies that report several miRNAs as important diagnostic and prognostic biomarkers in CM. Using publicly available NGS data, we analyzed the correlation of specific miRNA expression profiles with the expression signatures of known gene targets. Combining network analytics with machine learning, we developed specific non-linear classification models that could successfully predict CM recurrence and metastasis, based on two newly identified miRNA signatures. Subsequent unbiased analyses and independent test sets (i.e., a dataset not used for training, as a validation cohort) using our prediction models resulted in 73.85% and 82.09% accuracy in predicting CM recurrence and metastasis, respectively. Overall, our approach combines detailed analysis of miRNA profiles with heuristic optimization and machine learning, which facilitates dimensionality reduction and optimization of the prediction models. Our approach provides an improved prediction strategy that could serve as an auxiliary tool towards precision treatment. Full article
(This article belongs to the Special Issue Artificial Intelligence in Biomarker Discovery)
Show Figures

Figure 1

Back to TopTop