1
|
Wang C, Kumar GA, Rajapakse JC. Drug discovery and mechanism prediction with explainable graph neural networks. Sci Rep 2025; 15:179. [PMID: 39747341 PMCID: PMC11696803 DOI: 10.1038/s41598-024-83090-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Apprehension of drug action mechanism is paramount for drug response prediction and precision medicine. The unprecedented development of machine learning and deep learning algorithms has expedited the drug response prediction research. However, existing methods mainly focus on forward encoding of drugs, which is to obtain an accurate prediction of the response levels, but omitted to decipher the reaction mechanism between drug molecules and genes. We propose the eXplainable Graph-based Drug response Prediction (XGDP) approach that achieves a precise drug response prediction and reveals the comprehensive mechanism of action between drugs and their targets. XGDP represents drugs with molecular graphs, which naturally preserve the structural information of molecules and a Graph Neural Network module is applied to learn the latent features of molecules. Gene expression data from cancer cell lines are incorporated and processed by a Convolutional Neural Network module. A couple of deep learning attribution algorithms are leveraged to interpret interactions between drug molecular features and genes. We demonstrate that XGDP not only enhances the prediction accuracy compared to pioneering works but is also capable of capturing the salient functional groups of drugs and interactions with significant genes of cancer cells.
Collapse
Affiliation(s)
- Conghao Wang
- College of Computing and Data Science, Nanyang Technological University, Singapore, 639798, Singapore
| | - Gaurav Asok Kumar
- College of Computing and Data Science, Nanyang Technological University, Singapore, 639798, Singapore
| | - Jagath C Rajapakse
- College of Computing and Data Science, Nanyang Technological University, Singapore, 639798, Singapore.
| |
Collapse
|
2
|
Künzler T, Bamert M, Sprott H. Factors predicting treatment response to biological and targeted synthetic disease-modifying antirheumatic drugs in psoriatic arthritis - a systematic review and meta-analysis. Clin Rheumatol 2024; 43:3723-3746. [PMID: 39467905 PMCID: PMC11582271 DOI: 10.1007/s10067-024-07193-y] [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: 09/11/2024] [Revised: 10/07/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024]
Abstract
The therapeutic response of patients with psoriatic arthritis (PsA) varies greatly and is often unsatisfactory. Accordingly, it is essential to individualise treatment selection to minimise long-term complications. This study aimed to identify factors that might predict treatment response to biological and targeted synthetic disease-modifying antirheumatic drugs (bDMARDs and tsDMARDs) in patients with PsA and to outline their potential application using artificial intelligence (AI). Five electronic databases were screened to identify relevant studies. A random-effects meta-analysis was performed for factors that were investigated in at least four studies. Finally, 37 studies with a total of 17,042 patients were included. The most frequently investigated predictors in these studies were sex, age, C-reactive protein (CRP), the Health Assessment Questionnaire (HAQ), BMI, and disease duration. The meta-analysis revealed that male sex (odds ratio (OR) = 2.188, 95% confidence interval (CI) = 1.912-2.503) and higher baseline CRP (1.537, 1.111-2.125) were associated with greater treatment response. Older age (0.982, 0.975-0.99), higher baseline HAQ score (0.483, 0.336-0.696), higher baseline DAPSA score (0.789, 0.663-0.938), and higher baseline tender joint count (TJC) (0.97, 0.945-0.996) were negatively correlated with the response to therapy. The other factors were not statistically significant but might be of clinical importance in the context of a complex AI test battery. Further studies are needed to validate these findings and identify novel factors that could guide personalised treatment decisions for PsA patients, in particular in developing AI applications. In accordance with the latest medical developments, decision-support tools based on supervised learning algorithms have been proposed as a clinical application of these predictors. Key messages • Given the often unsatisfactory and unpredictable therapeutic response in patients with Psoriatic Arthritis (PsA), treatment selection must be highly individualized. • A systematic literature review was conducted to identify the most reliable predictors of treatment response to biologic and targeted synthetic disease-modifying antirheumatic drugs in PsA patients. • The potential integration of these predictors into AI tools for routine clinical practice is discussed.
Collapse
Affiliation(s)
- Tabea Künzler
- Medical Faculty of the University of Zurich, CH-8006, Zurich, Switzerland
| | - Manuel Bamert
- Retail Value Stream, Galenica AG, Untermattweg 8, CH-3027, Bern, Switzerland
| | - Haiko Sprott
- Medical Faculty of the University of Zurich, CH-8006, Zurich, Switzerland.
- Arztpraxis Hottingen, Hottingerstrasse 44, CH-8032, Zurich, Switzerland.
| |
Collapse
|
3
|
Wen J, Yang S, Zhu J, Liu A, Tan Q, Rao Y. Identifying feature genes of chickens with different feather pecking tendencies based on three machine learning algorithms and WGCNA. Front Vet Sci 2024; 11:1508397. [PMID: 39679174 PMCID: PMC11639596 DOI: 10.3389/fvets.2024.1508397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
Feather pecking (FP) is a significant welfare concern in poultry, which can result in reduced egg production, deterioration of feather condition, and an increase in mortality rate. This can harm the health of birds and the economic benefits of breeders. FP, as a complex trait, is regulated by multiple factors, and so far, no one has been able to elucidate its exact mechanism. In order to delve deeper into the genetic mechanism of FP, we acquired the expression matrix of dataset GSE36559. We analyzed the gene modules associated with the trait through WGCNA (Weighted correlation network analysis), and then used KEGG and GO to identify the biological pathways enriched by the modules using KEGG and GO. Subsequently, we analyzed the module with the highest correlation (0.99) using three machine learning (ML) algorithms to identify the feature genes that they collectively recognized. In this study, five feature genes, NUFIP2, ST14, OVM, GLULD1, and LOC424943, were identified. Finally, the discriminant value of the feature genes was evaluated by manipulating the receiver operating curve (ROC) in the external dataset GSE10380.
Collapse
Affiliation(s)
| | - Shenglin Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, Guizhou, China
| | | | | | | | | |
Collapse
|
4
|
Liu R, Zou Z, Zhang Z, He H, Xi M, Liang Y, Ye J, Dai Q, Wu Y, Tan H, Zhong W, Wang Z, Liang Y. Evaluation of glucocorticoid-related genes reveals GPD1 as a therapeutic target and regulator of sphingosine 1-phosphate metabolism in CRPC. Cancer Lett 2024; 605:217286. [PMID: 39413958 DOI: 10.1016/j.canlet.2024.217286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 09/08/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024]
Abstract
Prostate cancer (PCa) is an androgen-dependent disease, with castration-resistant prostate cancer (CRPC) being an advanced stage that no longer responds to androgen deprivation therapy (ADT). Mounting evidence suggests that glucocorticoid receptors (GR) confer resistance to ADT in CRPC patients by bypassing androgen receptor (AR) blockade. GR, as a novel therapeutic target in CRPC, has attracted substantial attention worldwide. This study utilized bioinformatic analysis of publicly available CRPC single-cell data to develop a consensus glucocorticoid-related signature (Glu-sig) that can serve as an independent predictor for relapse-free survival. Our results revealed that the signature demonstrated consistent and robust performance across seven publicly accessible datasets and an internal cohort. Furthermore, our findings demonstrated that glycerol-3-phosphate dehydrogenase 1 (GPD1) in Glu-sig can significantly promote CRPC progression by mediating the cell cycle pathway. Additionally, GPD1 was shown to be regulated by GR, with the GR antagonist mifepristone enhancing the anti-tumorigenic effects of GPD1 in CRPC cells. Mechanistically, targeting GPD1 induced the production of sphingosine 1-phosphate (S1P) and enhanced histone acetylation, thereby inducing the transcription of p21 that involved in cell cycle regulation. In conclusion, Glu-sig could serve as a robust and promising tool to improve the clinical outcomes of PCa patients, and modulating the GR/GPD1 axis that promotes tumor growth may be a promising approach for delaying CRPC progression.
Collapse
Affiliation(s)
- Ren Liu
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihao Zou
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China; Guangzhou Laboratory, Guangzhou, China
| | - Zhengrong Zhang
- Department of Urology, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Huichan He
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Ming Xi
- Department of Urology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Yingke Liang
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jianheng Ye
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qishan Dai
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongding Wu
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huijing Tan
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Weide Zhong
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China; Guangzhou Laboratory, Guangzhou, China; Macau Institute of Systems Engineering, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China.
| | - Zongren Wang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Yuxiang Liang
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Shao Y, Lv X, Ying S, Guo Q. Artificial Intelligence-Driven Precision Medicine: Multi-Omics and Spatial Multi-Omics Approaches in Diffuse Large B-Cell Lymphoma (DLBCL). FRONT BIOSCI-LANDMRK 2024; 29:404. [PMID: 39735973 DOI: 10.31083/j.fbl2912404] [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: 05/16/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 12/31/2024]
Abstract
In this comprehensive review, we delve into the transformative role of artificial intelligence (AI) in refining the application of multi-omics and spatial multi-omics within the realm of diffuse large B-cell lymphoma (DLBCL) research. We scrutinized the current landscape of multi-omics and spatial multi-omics technologies, accentuating their combined potential with AI to provide unparalleled insights into the molecular intricacies and spatial heterogeneity inherent to DLBCL. Despite current progress, we acknowledge the hurdles that impede the full utilization of these technologies, such as the integration and sophisticated analysis of complex datasets, the necessity for standardized protocols, the reproducibility of findings, and the interpretation of their biological significance. We proceeded to pinpoint crucial research voids and advocated for a trajectory that incorporates the development of advanced AI-driven data integration and analytical frameworks. The evolution of these technologies is crucial for enhancing resolution and depth in multi-omics studies. We also emphasized the importance of amassing extensive, meticulously annotated multi-omics datasets and fostering translational research efforts to connect laboratory discoveries with clinical applications seamlessly. Our review concluded that the synergistic integration of multi-omics, spatial multi-omics, and AI holds immense promise for propelling precision medicine forward in DLBCL. By surmounting the present challenges and steering towards the outlined futuristic pathways, we can harness these potent investigative tools to decipher the molecular and spatial conundrums of DLBCL. This will pave the way for refined diagnostic precision, nuanced risk stratification, and individualized therapeutic regimens, ushering in a new era of patient-centric oncology care.
Collapse
Affiliation(s)
- Yanping Shao
- Department of Hematology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009 Hangzhou, Zhejiang, China
| | - Xiuyan Lv
- Department of Hematology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 317000 Taizhou, Zhejiang, China
| | - Shuangwei Ying
- Department of Hematology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 317000 Taizhou, Zhejiang, China
| | - Qunyi Guo
- Department of Hematology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 317000 Taizhou, Zhejiang, China
| |
Collapse
|
6
|
Liu X, Shu X, Zhou Y, Jiang Y. Construction of a risk prediction model for postoperative deep vein thrombosis in colorectal cancer patients based on machine learning algorithms. Front Oncol 2024; 14:1499794. [PMID: 39664197 PMCID: PMC11631706 DOI: 10.3389/fonc.2024.1499794] [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: 09/21/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Background Colorectal cancer is a prevalent malignancy of the digestive system, with an increasing incidence. Lower extremity deep vein thrombosis (DVT) is a frequent postoperative complication, occurring in up to 40% of cases. Objective This research aims to develop and validate a machine learning model (ML) to predict the risk of lower limb deep vein thrombosis in patients with colorectal cancer, facilitating preventive and therapeutic measures to enhance recovery and ensure safety. Methods In this retrospective cohort study, we collected data from 429 colorectal cancer patients from January 2021 to January 2024. The medical records included age, blood test results, body mass index, underlying diseases, clinical staging, histological typing, surgical methods, and postoperative complications. We employed the Synthetic Minority Oversampling Technique to address imbalanced data and split the dataset into training and validation sets in a 7:3 ratio. Feature selection was performed using Random Forest (RF), XGBoost, and Least Absolute Shrinkage and Selection Operator algorithms (LASSO). We then trained six machine learning models: Logistic Regression (LR), Naive Bayes (NB), Gaussian Process (GP), Random Forest, XGBoost, and Multilayer Perceptron (MLP). The model's performance was evaluated using metrics such as area under the Receiver Operating Characteristic curve, accuracy, sensitivity, specificity, F1 score, and confusion matrix. Additionally, SHAP and LIME were used to enhance the interpretability of the results. Results The study combined Random Forest, XGBoost algorithms, and LASSO regression with univariate regression analysis to identify significant predictive factors, including age, preoperative prealbumin, preoperative albumin, preoperative hemoglobin, operation time, PIKVA2, CEA, and preoperative neutrophil count. The XGBoost model outperformed other ML algorithms, achieving an AUC of 0.996, an accuracy of 0.9636, a specificity of 0.9778, and an F1 score of 0.9576. Moreover, the SHAP method identified age and preoperative prealbumin as the primary determinants influencing ML model predictions. Finally, the study employed LIME for more precise prediction and interpretation of individual predictions. Conclusion The machine learning algorithms effectively predicted postoperative lower limb deep vein thrombosis in colorectal cancer patients. The XGBoost model demonstrated strong potential for improving early detection and treatment in clinical settings.
Collapse
Affiliation(s)
- Xin Liu
- Department of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Xingming Shu
- Department of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yejiang Zhou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yifan Jiang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
7
|
Kryczka J, Bachorz RA, Kryczka J, Boncela J. Radial Data Visualization-Based Step-by-Step Eliminative Algorithm to Predict Colorectal Cancer Patients' Response to FOLFOX Therapy. Int J Mol Sci 2024; 25:12149. [PMID: 39596218 PMCID: PMC11595261 DOI: 10.3390/ijms252212149] [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: 09/16/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Application of the FOLFOX scheme to colorectal cancer (CRC) patients often results in the development of chemo-resistance, leading to therapy failure. This study aimed to develop a functional and easy-to-use algorithm to predict patients' response to FOLFOX treatment. Transcriptomic data of CRC patient's samples treated with FOLFOX were downloaded from the Gene Expression Omnibus database (GSE83129, GSE28702, GSE69657, GSE19860 and GSE41568). Comparing the expression of top up- and downregulated genes in FOLFOX responder and non-responder patients' groups, we selected 30 potential markers that were used to create a step-by-step eliminative procedure based on modified radial data visualization, which depicts the interplay between the expression level of chosen attributes (genes) to locate data points in low-dimensional space. Our analysis proved that FOLFOX-resistant CRC samples are predominantly characterized by upregulated expression levels of TMEM182 and MCM9 and downregulated LRRFIP1. Additionally, the procedure developed based on expression levels of TMEM182, MCM9, LRRFIP1, LAMP1, FAM161A, KLHL36, ETV5, RNF168, SRSF11, NCKAP5, CRTAP, VAMP2, ZBTB49 and RIMBP2 proved to be capable in predicting FOLFOX therapy response. In conclusion, our approach can give a unique insight into clinical decision-making regarding therapy scheme administration, potentially increasing patients' survival and, consequently, medical futility due to incorrect therapy application.
Collapse
Affiliation(s)
- Jakub Kryczka
- Laboratory of Cell Signaling, Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa St., 93-232 Lodz, Poland;
| | - Rafał Adam Bachorz
- Laboratory of Molecular Modeling, Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa St., 93-232 Lodz, Poland;
| | - Jolanta Kryczka
- Department of Biomedicine and Genetics, Medical University of Lodz, 92-213 Lodz, Poland;
| | - Joanna Boncela
- Laboratory of Cell Signaling, Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa St., 93-232 Lodz, Poland;
| |
Collapse
|
8
|
Sahib NRBM, Mohamed JS, Rashid MBMA, Jayalakshmi, Lin YC, Chee YL, Fan BE, De Mel S, Ooi MGM, Jen WY, Chow EKH. A Combinatorial Functional Precision Medicine Platform for Rapid Therapeutic Response Prediction in AML. Cancer Med 2024; 13:e70401. [PMID: 39560206 PMCID: PMC11574777 DOI: 10.1002/cam4.70401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Despite advances made in targeted biomarker-based therapy for acute myeloid leukemia (AML) treatment, remission is often short and followed by relapse and acquired resistance. Functional precision medicine (FPM) efforts have been shown to improve therapy selection guidance by incorporating comprehensive biological data to tailor individual treatment. However, effectively managing complex biological data, while also ensuring rapid conversion of actionable insights into clinical utility remains challenging. METHODS We have evaluated the clinical applicability of quadratic phenotypic optimization platform (QPOP), to predict clinical response to combination therapies in AML and reveal patient-centric insights into combination therapy sensitivities. In this prospective study, 51 primary samples from newly diagnosed (ND) or refractory/relapsed (R/R) AML patients were evaluated by QPOP following ex vivo drug testing. RESULTS Individualized drug sensitivity reports were generated in 55/63 (87.3%) patient samples with a median turnaround time of 5 (4-10) days from sample collection to report generation. To evaluate clinical feasibility, QPOP-predicted response was compared to clinical treatment outcomes and indicated concordant results with 83.3% sensitivity and 90.9% specificity and an overall 86.2% accuracy. Serial QPOP analysis in a FLT3-mutant patient sample indicated decreased FLT3 inhibitor (FLT3i) sensitivity, which is concordant with increasing FLT3 allelic burden and drug resistance development. Forkhead box M1 (FOXM1)-AKT signaling was subsequently identified to contribute to resistance to FLT3i. CONCLUSION Overall, this study demonstrates the feasibility of applying QPOP as a functional combinatorial precision medicine platform to predict therapeutic sensitivities in AML and provides the basis for prospective clinical trials evaluating ex vivo-guided combination therapy.
Collapse
Affiliation(s)
- Noor Rashidha Binte Meera Sahib
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jameelah Sheik Mohamed
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | | | - Jayalakshmi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Yen Lin Chee
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Bingwen Eugene Fan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology, Tan Tock Seng Hospital, Singapore
- Lee Kong Chain School of Medicine, Nanyang Technological University, Singapore
- Department of Laboratory Medicine, Khoo Teck Puat Hospital, Singapore
| | - Sanjay De Mel
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Melissa Gaik Ming Ooi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Wei-Ying Jen
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore
| |
Collapse
|
9
|
Zhao Y, Ma X, Meng X, Li H, Tang Q. Integrating machine learning and single-cell transcriptomic analysis to identify potential biomarkers and analyze immune features of ischemic stroke. Sci Rep 2024; 14:26069. [PMID: 39478056 PMCID: PMC11525974 DOI: 10.1038/s41598-024-77495-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
This study employs machine learning and single-cell transcriptome sequencing (scRNA-seq) analysis to unearth novel biomarkers and delineate the immune characteristics of ischemic stroke (IS), thereby contributing fresh insights into IS treatment strategies.Our research leverages gene expression data sourced from the GEO database. We undertake weighted gene co-expression network analysis (WGCNA) to filter pertinent genes and subsequently employ machine learning algorithms for the identification of feature genes. Concurrently, we rigorously execute quality control measures, dimensionality reduction techniques, and cell annotation on the scRNA-seq data to pinpoint differentially expressed genes (DEGs). The identification of core genes, denoted as Hub genes, among the feature genes and DEGs, is achieved through meticulous overlapping analysis. We illuminate the immune characteristics of these Hub genes using a suite of analytical tools, encompassing CIBERSORT, MCPcounter, and pseudotemporal analysis, all based on immune cell annotations and single-cell transcriptome data.Subsequently, we harness the CMap database to prognosticate potential therapeutic drugs and scrutinize their associations with the identified Hub genes. Our findings unveil robust linkages between three pivotal Hub genes-namely, RNF13, VASP, and CD163-and specific immune cell types such as T cells and neutrophils. These Hub genes predominantly manifest in macrophages and microglial cells within the scRNA-seq immune cell population, exhibiting variances across different stages of cellular differentiation. In conclusion, this study unearths highly pertinent biomarkers for IS diagnosis and elucidates IS-induced immune infiltration characteristics, thus providing a firm foundation for a comprehensive exploration of potential immune mechanisms and the identification of novel therapeutic targets for IS.
Collapse
Affiliation(s)
- Yaowei Zhao
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Xiyuan Ma
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Xianghong Meng
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Hongyu Li
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China.
- Second Affiliated Hospital of Heilongjiang, University of Chinese Medicine, Harbin, 150000, Heilongjiang, China.
| | - Qiang Tang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China.
- Second Affiliated Hospital of Heilongjiang, University of Chinese Medicine, Harbin, 150000, Heilongjiang, China.
| |
Collapse
|
10
|
De Landtsheer S, Badkas A, Kulms D, Sauter T. Model ensembling as a tool to form interpretable multi-omic predictors of cancer pharmacosensitivity. Brief Bioinform 2024; 25:bbae567. [PMID: 39494610 PMCID: PMC11532660 DOI: 10.1093/bib/bbae567] [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: 07/10/2024] [Revised: 09/23/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Stratification of patients diagnosed with cancer has become a major goal in personalized oncology. One important aspect is the accurate prediction of the response to various drugs. It is expected that the molecular characteristics of the cancer cells contain enough information to retrieve specific signatures, allowing for accurate predictions based solely on these multi-omic data. Ideally, these predictions should be explainable to clinicians, in order to be integrated in the patients care. We propose a machine-learning framework based on ensemble learning to integrate multi-omic data and predict sensitivity to an array of commonly used and experimental compounds, including chemotoxic compounds and targeted kinase inhibitors. We trained a set of classifiers on the different parts of our dataset to produce omic-specific signatures, then trained a random forest classifier on these signatures to predict drug responsiveness. We used the Cancer Cell Line Encyclopedia dataset, comprising multi-omic and drug sensitivity measurements for hundreds of cell lines, to build the predictive models, and validated the results using nested cross-validation. Our results show good performance for several compounds (Area under the Receiver-Operating Curve >79%) across the most frequent cancer types. Furthermore, the simplicity of our approach allows to examine which omic layers have a greater importance in the models and identify new putative markers of drug responsiveness. We propose several models based on small subsets of transcriptional markers with the potential to become useful tools in personalized oncology, paving the way for clinicians to use the molecular characteristics of the tumors to predict sensitivity to therapeutic compounds.
Collapse
Affiliation(s)
- Sébastien De Landtsheer
- Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l’Université, L4365 Esch-sur-Alzette, Luxembourg
| | - Apurva Badkas
- Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l’Université, L4365 Esch-sur-Alzette, Luxembourg
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, Technische Universität-Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases, Technische Universität-Dresden, 01307 Dresden, Germany
| | - Thomas Sauter
- Department of Life Sciences and Medicine, University of Luxembourg, 2, place de l’Université, L4365 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
11
|
Xu F, Lai J. Commentary: Immune cell infiltration and prognostic index in cervical cancer: insights from metabolism-related differential genes. Front Immunol 2024; 15:1446741. [PMID: 39364407 PMCID: PMC11446798 DOI: 10.3389/fimmu.2024.1446741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024] Open
Affiliation(s)
- Fangshi Xu
- Department of Vascular Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiawei Lai
- Department of Urology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
12
|
Froicu EM, Oniciuc OM, Afrăsânie VA, Marinca MV, Riondino S, Dumitrescu EA, Alexa-Stratulat T, Radu I, Miron L, Bacoanu G, Poroch V, Gafton B. The Use of Artificial Intelligence in Predicting Chemotherapy-Induced Toxicities in Metastatic Colorectal Cancer: A Data-Driven Approach for Personalized Oncology. Diagnostics (Basel) 2024; 14:2074. [PMID: 39335752 PMCID: PMC11431340 DOI: 10.3390/diagnostics14182074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 08/31/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Machine learning models learn about general behavior from data by finding the relationships between features. Our purpose was to develop a predictive model to identify and predict which subset of colorectal cancer patients are more likely to experience chemotherapy-induced toxicity and to determine the specific attributes that influence the presence of treatment-related side effects. METHODS The predictor was general toxicity, and for the construction of our data training, we selected 95 characteristics that represent the health state of 74 patients prior to their first round of chemotherapy. After the data were processed, Random Forest models were trained to offer an optimal balance between accuracy and interpretability. RESULTS We constructed a machine learning predictor with an emphasis on assessing the importance of numerical and categorical variables in relation to toxicity. CONCLUSIONS The incorporation of artificial intelligence in personalizing colorectal cancer management by anticipating and overseeing toxicities more effectively illustrates a pivotal shift towards more personalized and precise medical care.
Collapse
Affiliation(s)
- Eliza-Maria Froicu
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania
- Department of Oncology, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- 2nd Internal Medicine Department, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Oriana-Maria Oniciuc
- Faculty of Computer Science, "Alexandru Ioan Cuza" University, 700506 Iasi, Romania
| | - Vlad-Adrian Afrăsânie
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania
- Department of Oncology, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihai-Vasile Marinca
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania
- Department of Oncology, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Silvia Riondino
- Department of Systems Medicine, Medical Oncology, Tor Vergata Clinical Center, University of Rome "Tor Vergata", Viale Oxford 81, 00133 Rome, Italy
| | - Elena Adriana Dumitrescu
- Department of Oncology, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Institute of Oncology Prof. Dr. Alexandru Trestioreanu, Șoseaua Fundeni, 022328 Bucharest, Romania
| | - Teodora Alexa-Stratulat
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania
- Department of Oncology, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Iulian Radu
- First Surgical Oncology Unit, Department of Surgery, Regional Institute of Oncology, 700483 Iasi, Romania
- Department of Surgery, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Lucian Miron
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania
- Department of Oncology, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Gema Bacoanu
- 2nd Internal Medicine Department, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Palliative Care, Regional Institute of Oncology, 700483 Iasi, Romania
| | - Vladimir Poroch
- 2nd Internal Medicine Department, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Palliative Care, Regional Institute of Oncology, 700483 Iasi, Romania
| | - Bogdan Gafton
- Department of Medical Oncology, Regional Institute of Oncology, 700483 Iasi, Romania
- Department of Oncology, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
13
|
Kamble P, Nagar PR, Bhakhar KA, Garg P, Sobhia ME, Naidu S, Bharatam PV. Cancer pharmacoinformatics: Databases and analytical tools. Funct Integr Genomics 2024; 24:166. [PMID: 39294509 DOI: 10.1007/s10142-024-01445-5] [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: 07/29/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Cancer is a subject of extensive investigation, and the utilization of omics technology has resulted in the generation of substantial volumes of big data in cancer research. Numerous databases are being developed to manage and organize this data effectively. These databases encompass various domains such as genomics, transcriptomics, proteomics, metabolomics, immunology, and drug discovery. The application of computational tools into various core components of pharmaceutical sciences constitutes "Pharmacoinformatics", an emerging paradigm in rational drug discovery. The three major features of pharmacoinformatics include (i) Structure modelling of putative drugs and targets, (ii) Compilation of databases and analysis using statistical approaches, and (iii) Employing artificial intelligence/machine learning algorithms for the discovery of novel therapeutic molecules. The development, updating, and analysis of databases using statistical approaches play a pivotal role in pharmacoinformatics. Multiple software tools are associated with oncoinformatics research. This review catalogs the databases and computational tools related to cancer drug discovery and highlights their potential implications in the pharmacoinformatics of cancer.
Collapse
Affiliation(s)
- Pradnya Kamble
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Prinsa R Nagar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Kaushikkumar A Bhakhar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - M Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Srivatsava Naidu
- Center of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Prasad V Bharatam
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India.
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India.
| |
Collapse
|
14
|
Lv F, Li X, Wang Z, Wang X, Liu J. Identification and validation of Rab GTPases RAB13 as biomarkers for peritoneal metastasis and immune cell infiltration in colorectal cancer patients. Front Immunol 2024; 15:1403008. [PMID: 39192986 PMCID: PMC11347351 DOI: 10.3389/fimmu.2024.1403008] [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: 03/18/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Background As one of the most common cancer, colorectal cancer (CRC) is with high morbidity and mortality. Peritoneal metastasis (PM) is a fatal state of CRC, and few patients may benefit from traditional therapies. There is a complex interaction between PM and immune cell infiltration. Therefore, we aimed to determine biomarkers associated with colorectal cancer peritoneal metastasis (CRCPM) and their relationship with immune cell infiltration. Methods By informatic analysis, differently expressed genes (DEGs) were selected and hub genes were screened out. RAB13, one of the hub genes, was identificated from public databases and validated in CRC tissues. The ESTIMATE, CEBERSORT and TIMER algorithms were applied to analyze the correlation between RAB13 and immune infiltration in CRC. RAB13's expression in different cells were analyzed at the single-cell level in scRNA-Seq. The Gene Set Enrichment Analysis (GSEA) was performed for RAB13 enrichment and further confirmed. Using oncoPredict algorithm, RAB13's impact on drug sensitivity was evaluated. Results High RAB13 expression was identified in public databases and led to a poor prognosis. RAB13 was found to be positively correlated with the macrophages and other immune cells infiltration and from scRNA-Seq, RAB13 was found to be located in CRC cells and macrophages. GSEA revealed that high RAB13 expression enriched in a various of biological signaling, and oncoPredict algorithm showed that RAB13 expression was correlated with paclitaxel sensitivity. Conclusion Our study indicated clinical role of RAB13 in CRC-PM, suggesting its potential as a therapeutic target in the future.
Collapse
Affiliation(s)
- Fei Lv
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaoqi Li
- Oncology Department III, People’s Hospital of Liaoning Province, Shenyang, Liaoning, China
| | - Zhe Wang
- Department of Digestive Diseases 1, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Xiaobo Wang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jing Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
15
|
Gültepe Y, Berber S, Gültepe N. Modeling and predicting meat yield and growth performance using morphological features of narrow-clawed crayfish with machine learning techniques. Sci Rep 2024; 14:18499. [PMID: 39122763 PMCID: PMC11315914 DOI: 10.1038/s41598-024-69539-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024] Open
Abstract
In recent studies, artificial intelligence and machine learning methods give higher accuracy than other prediction methods in large data sets with complex structures. Instead of statistical methods, artificial intelligence, and machine learning are used due to the difficulty of constructing mathematical models in multi-parameter and multivariate problems. In this study, predictions of length-weight relationships and meat productivity were generated by machine learning models using measurement data of male and female crayfish in the narrow-clawed crayfish population living in Apolyont Lake. The data set was created using the growth performance and morphometric characters from 1416 crayfish in different years to determine the length-weight relationship and length-meat yield. Statistical methods, artificial intelligence, and machine learning are used due to the difficulty of constructing mathematical models in multi-parameter and multivariate problems. The analysis results show that most models designed as an alternative to traditional estimation methods in future planning studies in sustainable fisheries, aquaculture, and natural sources management are valid for machine learning and artificial intelligence. Seven different machine learning algorithms were applied to the data set and the length-weight relationships and length-meat yields were evaluated for both male and female individuals. Support vector regression (SVR) has achieved the best prediction performance accuracy with 0.996 and 0.992 values for the length-weight of males and females, with 0.996 and 0.995 values for the length-meat yield of males and females. The results showed that the SVR outperforms the others for all scenarios regarding the accuracy, sensitivity, and specificity metrics.
Collapse
Affiliation(s)
- Yasemin Gültepe
- Faculty of Engineering, Department of Software Engineering, Atatürk University, 25240, Erzurum, Türkiye
| | - Selçuk Berber
- Faculty of Marine Science and Technology, Department of Fisheries Fundamental Sciences, Çanakkale Onsekiz Mart University, 17100, Çanakkale, Türkiye
| | - Nejdet Gültepe
- Faculty of Fisheries, Department of Fisheries Fundamental Sciences, Atatürk University, 25240, Erzurum, Türkiye.
| |
Collapse
|
16
|
Meerwijk EL, McElfresh DC, Martins S, Tamang SR. Evaluating accuracy and fairness of clinical decision support algorithms when health care resources are limited. J Biomed Inform 2024; 156:104664. [PMID: 38851413 DOI: 10.1016/j.jbi.2024.104664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/02/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE Guidance on how to evaluate accuracy and algorithmic fairness across subgroups is missing for clinical models that flag patients for an intervention but when health care resources to administer that intervention are limited. We aimed to propose a framework of metrics that would fit this specific use case. METHODS We evaluated the following metrics and applied them to a Veterans Health Administration clinical model that flags patients for intervention who are at risk of overdose or a suicidal event among outpatients who were prescribed opioids (N = 405,817): Receiver - Operating Characteristic and area under the curve, precision - recall curve, calibration - reliability curve, false positive rate, false negative rate, and false omission rate. In addition, we developed a new approach to visualize false positives and false negatives that we named 'per true positive bars.' We demonstrate the utility of these metrics to our use case for three cohorts of patients at the highest risk (top 0.5 %, 1.0 %, and 5.0 %) by evaluating algorithmic fairness across the following age groups: <=30, 31-50, 51-65, and >65 years old. RESULTS Metrics that allowed us to assess group differences more clearly were the false positive rate, false negative rate, false omission rate, and the new 'per true positive bars'. Metrics with limited utility to our use case were the Receiver - Operating Characteristic and area under the curve, the calibration - reliability curve, and the precision - recall curve. CONCLUSION There is no "one size fits all" approach to model performance monitoring and bias analysis. Our work informs future researchers and clinicians who seek to evaluate accuracy and fairness of predictive models that identify patients to intervene on in the context of limited health care resources. In terms of ease of interpretation and utility for our use case, the new 'per true positive bars' may be the most intuitive to a range of stakeholders and facilitates choosing a threshold that allows weighing false positives against false negatives, which is especially important when predicting severe adverse events.
Collapse
Affiliation(s)
- Esther L Meerwijk
- Program Evaluation and Resource Center, Office of Mental Health and Suicide Prevention, Department of Veterans Affairs, Menlo Park, CA, USA; VA Health Systems Research, Center for Innovation to Implementation (Ci2i), VA Palo Alto Health Care System, Menlo Park, CA, USA.
| | - Duncan C McElfresh
- Program Evaluation and Resource Center, Office of Mental Health and Suicide Prevention, Department of Veterans Affairs, Menlo Park, CA, USA
| | - Susana Martins
- Program Evaluation and Resource Center, Office of Mental Health and Suicide Prevention, Department of Veterans Affairs, Menlo Park, CA, USA
| | - Suzanne R Tamang
- Program Evaluation and Resource Center, Office of Mental Health and Suicide Prevention, Department of Veterans Affairs, Menlo Park, CA, USA; VA Health Systems Research, Center for Innovation to Implementation (Ci2i), VA Palo Alto Health Care System, Menlo Park, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
17
|
Mohammadzadeh-Vardin T, Ghareyazi A, Gharizadeh A, Abbasi K, Rabiee HR. DeepDRA: Drug repurposing using multi-omics data integration with autoencoders. PLoS One 2024; 19:e0307649. [PMID: 39058696 PMCID: PMC11280260 DOI: 10.1371/journal.pone.0307649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer treatment has become one of the biggest challenges in the world today. Different treatments are used against cancer; drug-based treatments have shown better results. On the other hand, designing new drugs for cancer is costly and time-consuming. Some computational methods, such as machine learning and deep learning, have been suggested to solve these challenges using drug repurposing. Despite the promise of classical machine-learning methods in repurposing cancer drugs and predicting responses, deep-learning methods performed better. This study aims to develop a deep-learning model that predicts cancer drug response based on multi-omics data, drug descriptors, and drug fingerprints and facilitates the repurposing of drugs based on those responses. To reduce multi-omics data's dimensionality, we use autoencoders. As a multi-task learning model, autoencoders are connected to MLPs. We extensively tested our model using three primary datasets: GDSC, CTRP, and CCLE to determine its efficacy. In multiple experiments, our model consistently outperforms existing state-of-the-art methods. Compared to state-of-the-art models, our model achieves an impressive AUPRC of 0.99. Furthermore, in a cross-dataset evaluation, where the model is trained on GDSC and tested on CCLE, it surpasses the performance of three previous works, achieving an AUPRC of 0.72. In conclusion, we presented a deep learning model that outperforms the current state-of-the-art regarding generalization. Using this model, we could assess drug responses and explore drug repurposing, leading to the discovery of novel cancer drugs. Our study highlights the potential for advanced deep learning to advance cancer therapeutic precision.
Collapse
Affiliation(s)
- Taha Mohammadzadeh-Vardin
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| | - Amin Ghareyazi
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| | - Ali Gharizadeh
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| | - Karim Abbasi
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
- Faculty of Mathematics and Computer Science, Kharazmi University, Tehran, Iran
| | - Hamid R. Rabiee
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
18
|
Park S, Silva E, Singhal A, Kelly MR, Licon K, Panagiotou I, Fogg C, Fong S, Lee JJY, Zhao X, Bachelder R, Parker BA, Yeung KT, Ideker T. A deep learning model of tumor cell architecture elucidates response and resistance to CDK4/6 inhibitors. NATURE CANCER 2024; 5:996-1009. [PMID: 38443662 PMCID: PMC11286358 DOI: 10.1038/s43018-024-00740-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 02/07/2024] [Indexed: 03/07/2024]
Abstract
Cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6is) have revolutionized breast cancer therapy. However, <50% of patients have an objective response, and nearly all patients develop resistance during therapy. To elucidate the underlying mechanisms, we constructed an interpretable deep learning model of the response to palbociclib, a CDK4/6i, based on a reference map of multiprotein assemblies in cancer. The model identifies eight core assemblies that integrate rare and common alterations across 90 genes to stratify palbociclib-sensitive versus palbociclib-resistant cell lines. Predictions translate to patients and patient-derived xenografts, whereas single-gene biomarkers do not. Most predictive assemblies can be shown by CRISPR-Cas9 genetic disruption to regulate the CDK4/6i response. Validated assemblies relate to cell-cycle control, growth factor signaling and a histone regulatory complex that we show promotes S-phase entry through the activation of the histone modifiers KAT6A and TBL1XR1 and the transcription factor RUNX1. This study enables an integrated assessment of how a tumor's genetic profile modulates CDK4/6i resistance.
Collapse
Affiliation(s)
- Sungjoon Park
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Erica Silva
- Program in Biomedical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Akshat Singhal
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Marcus R Kelly
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, San Diego, CA, USA
| | - Kate Licon
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Isabella Panagiotou
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Catalina Fogg
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Samson Fong
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - John J Y Lee
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Xiaoyu Zhao
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Robin Bachelder
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Barbara A Parker
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, San Diego, CA, USA
| | - Kay T Yeung
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California, San Diego, San Diego, CA, USA
| | - Trey Ideker
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California, San Diego, San Diego, CA, USA.
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
19
|
Lenhof K, Eckhart L, Rolli LM, Volkamer A, Lenhof HP. Reliable anti-cancer drug sensitivity prediction and prioritization. Sci Rep 2024; 14:12303. [PMID: 38811639 PMCID: PMC11137046 DOI: 10.1038/s41598-024-62956-6] [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: 11/28/2023] [Accepted: 05/23/2024] [Indexed: 05/31/2024] Open
Abstract
The application of machine learning (ML) to solve real-world problems does not only bear great potential but also high risk. One fundamental challenge in risk mitigation is to ensure the reliability of the ML predictions, i.e., the model error should be minimized, and the prediction uncertainty should be estimated. Especially for medical applications, the importance of reliable predictions can not be understated. Here, we address this challenge for anti-cancer drug sensitivity prediction and prioritization. To this end, we present a novel drug sensitivity prediction and prioritization approach guaranteeing user-specified certainty levels. The developed conformal prediction approach is applicable to classification, regression, and simultaneous regression and classification. Additionally, we propose a novel drug sensitivity measure that is based on clinically relevant drug concentrations and enables a straightforward prioritization of drugs for a given cancer sample.
Collapse
Affiliation(s)
- Kerstin Lenhof
- Center for Bioinformatics, Chair for Bioinformatics, Saarland Informatics Campus (E2.1) Saarland University, Campus, 66123, Saarbrücken, Saarland, Germany.
| | - Lea Eckhart
- Center for Bioinformatics, Chair for Bioinformatics, Saarland Informatics Campus (E2.1) Saarland University, Campus, 66123, Saarbrücken, Saarland, Germany
| | - Lisa-Marie Rolli
- Center for Bioinformatics, Chair for Bioinformatics, Saarland Informatics Campus (E2.1) Saarland University, Campus, 66123, Saarbrücken, Saarland, Germany
| | - Andrea Volkamer
- Center for Bioinformatics, Chair for Data Driven Drug Design, Saarland Informatics Campus (E2.1) Saarland University, Campus, 66123, Saarbrücken, Saarland, Germany
| | - Hans-Peter Lenhof
- Center for Bioinformatics, Chair for Bioinformatics, Saarland Informatics Campus (E2.1) Saarland University, Campus, 66123, Saarbrücken, Saarland, Germany
| |
Collapse
|
20
|
Su L, Hounye AH, Pan Q, Miao K, Wang J, Hou M, Xiong L. Explainable cancer factors discovery: Shapley additive explanation for machine learning models demonstrates the best practices in the case of pancreatic cancer. Pancreatology 2024; 24:404-423. [PMID: 38342661 DOI: 10.1016/j.pan.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/07/2024] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
Pancreatic cancer is one of digestive tract cancers with high mortality rate. Despite the wide range of available treatments and improvements in surgery, chemotherapy, and radiation therapy, the five-year prognosis for individuals diagnosed pancreatic cancer remains poor. There is still research to be done to see if immunotherapy may be used to treat pancreatic cancer. The goals of our research were to comprehend the tumor microenvironment of pancreatic cancer, found a useful biomarker to assess the prognosis of patients, and investigated its biological relevance. In this paper, machine learning methods such as random forest were fused with weighted gene co-expression networks for screening hub immune-related genes (hub-IRGs). LASSO regression model was used to further work. Thus, we got eight hub-IRGs. Based on hub-IRGs, we created a prognosis risk prediction model for PAAD that can stratify accurately and produce a prognostic risk score (IRG_Score) for each patient. In the raw data set and the validation data set, the five-year area under the curve (AUC) for this model was 0.9 and 0.7, respectively. And shapley additive explanation (SHAP) portrayed the importance of prognostic risk prediction influencing factors from a machine learning perspective to obtain the most influential certain gene (or clinical factor). The five most important factors were TRIM67, CORT, PSPN, SCAMP5, RFXAP, all of which are genes. In summary, the eight hub-IRGs had accurate risk prediction performance and biological significance, which was validated in other cancers. The result of SHAP helped to understand the molecular mechanism of pancreatic cancer.
Collapse
Affiliation(s)
- Liuyan Su
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China
| | | | - Qi Pan
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China
| | - Kexin Miao
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China
| | - Jiaoju Wang
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China
| | - Muzhou Hou
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China.
| | - Li Xiong
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Clinical Research Center for Intelligent General Surgery, Changsha, 410011, China.
| |
Collapse
|
21
|
Ovchinnikova K, Born J, Chouvardas P, Rapsomaniki M, Kruithof-de Julio M. Overcoming limitations in current measures of drug response may enable AI-driven precision oncology. NPJ Precis Oncol 2024; 8:95. [PMID: 38658785 PMCID: PMC11043358 DOI: 10.1038/s41698-024-00583-0] [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: 08/16/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Machine learning (ML) models of drug sensitivity prediction are becoming increasingly popular in precision oncology. Here, we identify a fundamental limitation in standard measures of drug sensitivity that hinders the development of personalized prediction models - they focus on absolute effects but do not capture relative differences between cancer subtypes. Our work suggests that using z-scored drug response measures mitigates these limitations and leads to meaningful predictions, opening the door for sophisticated ML precision oncology models.
Collapse
Affiliation(s)
- Katja Ovchinnikova
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Panagiotis Chouvardas
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Marianna Kruithof-de Julio
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland.
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
22
|
You C, Ren J, Cheng L, Peng C, Lu P, Guo K, Zhong F, Wang J, Gao X, Cao J, Liu H, Liu T. Development and validation of a machine learning-based postoperative prognostic model for plasma cell neoplasia with spinal lesions as initial clinical manifestations: a single-center cohort study. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2024:10.1007/s00586-024-08223-8. [PMID: 38584243 DOI: 10.1007/s00586-024-08223-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/13/2024] [Accepted: 03/07/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Spinal multiple myeloma (MM) and solitary plasmacytoma of bone (SPB), both plasma cell neoplasms, greatly affect patients' quality of life due to spinal involvement. Accurate prediction of surgical outcomes is crucial for personalized patient care, but systematic treatment guidelines and predictive models are lacking. OBJECTIVE This study aimed to develop and validate a machine learning (ML)-based model to predict postoperative outcomes and identify prognostic factors for patients with spinal MM and SPB. METHODS A retrospective analysis was conducted on patients diagnosed with MM or SPB from 2011 to 2015, followed by prospective data collection from 2016 to 2017. Patient demographics, tumor characteristics, clinical treatments, and laboratory results were analyzed as input features. Four types of ML algorithms were employed for model development. The performance was assessed using discrimination and calibration measures, and the Shapley Additive exPlanations (SHAP) method was applied for model interpretation. RESULTS A total of 169 patients were included, with 119 for model training and 50 for validation. The Gaussian Naïve Bayes (GNB) model exhibited superior predictive accuracy and stability. Prospective validation on the 50 patients revealed an area under the curve (AUC) of 0.863, effectively distinguishing between 5-year survivors and non-survivors. Key prognostic factors identified included International Staging System (ISS) stage, Durie-Salmon (DS) stage, targeted therapy, and age. CONCLUSIONS The GNB model has the best performance and high reliability in predicting postoperative outcomes. Variables such as ISS stage and DS stage were significant in influencing patient prognosis. This study enhances the ability to identify patients at risk of poor outcomes, thereby aiding clinical decision-making.
Collapse
Affiliation(s)
- Chaoqun You
- Department of Joint Surgery, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, 261061, China
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Jiaji Ren
- Department of Joint Surgery, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, 261061, China
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Linfei Cheng
- Department of Joint Surgery, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, 261061, China
- School of Medicine, Anhui University of Science and Technology, No.168 Taifeng Road, Huainan, 232001, China
| | - Cheng Peng
- Department of Joint Surgery, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, 261061, China
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Peng Lu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200003, China
| | - Kai Guo
- Department of Orthopedics, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, No. 164 Lanxi Road, Shanghai, 200062, China
| | - Fulong Zhong
- Department of Joint Surgery, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, 261061, China
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Jing Wang
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Xin Gao
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Jiashi Cao
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai, 200003, China.
- Department of Orthopedics, No. 455 Hospital of the Chinese People's Liberation Army, The Navy Medical University, No. 338 Huaihai West Road, Shanghai, 200052, China.
| | - Huancai Liu
- Department of Joint Surgery, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, 261061, China.
| | - Tielong Liu
- Department of Joint Surgery, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, 261061, China.
- Department of Orthopaedic Oncology, Changzheng Hospital of the Navy Medical University, No. 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
23
|
Chen L, Hua J, He X. Bioinformatics analysis identifies a key gene HLA_DPA1 in severe influenza-associated immune infiltration. BMC Genomics 2024; 25:257. [PMID: 38454348 PMCID: PMC10918912 DOI: 10.1186/s12864-024-10184-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 03/04/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Severe influenza is a serious global health issue that leads to prolonged hospitalization and mortality on a significant scale. The pathogenesis of this infectious disease is poorly understood. Therefore, this study aimed to identify the key genes associated with severe influenza patients necessitating invasive mechanical ventilation. METHODS The current study utilized two publicly accessible gene expression profiles (GSE111368 and GSE21802) from the Gene Expression Omnibus database. The research focused on identifying the genes exhibiting differential expression between severe and non-severe influenza patients. We employed three machine learning algorithms, namely the Least Absolute Shrinkage and Selection Operator regression model, Random Forest, and Support Vector Machine-Recursive Feature Elimination, to detect potential key genes. The key gene was further selected based on the diagnostic performance of the target genes substantiated in the dataset GSE101702. A single-sample gene set enrichment analysis algorithm was applied to evaluate the participation of immune cell infiltration and their associations with key genes. RESULTS A total of 44 differentially expressed genes were recognized; among them, we focused on 10 common genes, namely PCOLCE2, HLA_DPA1, LOC653061, TDRD9, MPO, HLA_DQA1, MAOA, S100P, RAP1GAP, and CA1. To ensure the robustness of our findings, we employed overlapping LASSO regression, Random Forest, and SVM-RFE algorithms. By utilizing these algorithms, we were able to pinpoint the aforementioned 10 genes as potential biomarkers for distinguishing between both cases of influenza (severe and non-severe). However, the gene HLA_DPA1 has been recognized as a crucial factor in the pathological condition of severe influenza. Notably, the validation dataset revealed that this gene exhibited the highest area under the receiver operating characteristic curve, with a value of 0.891. The use of single-sample gene set enrichment analysis has provided valuable insights into the immune responses of patients afflicted with severe influenza that have further revealed a categorical correlation between the expression of HLA_DPA1 and lymphocytes. CONCLUSION The findings indicated that the HLA_DPA1 gene may play a crucial role in the immune-pathological condition of severe influenza and could serve as a promising therapeutic target for patients infected with severe influenza.
Collapse
Affiliation(s)
- Liang Chen
- Department of Infectious Diseases, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical College of Nanjing University, No 188, Lingshan North Road, Qixia District, Nanjing, 210046, China.
| | - Jie Hua
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaopu He
- Department of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Ramović Hamzagić A, Cvetković D, Gazdić Janković M, Milivojević Dimitrijević N, Nikolić D, Živanović M, Kastratović N, Petrović I, Nikolić S, Jovanović M, Šeklić D, Filipović N, Ljujić B. Modeling 5-FU-Induced Chemotherapy Selection of a Drug-Resistant Cancer Stem Cell Subpopulation. Curr Oncol 2024; 31:1221-1234. [PMID: 38534924 PMCID: PMC10968802 DOI: 10.3390/curroncol31030091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: Cancer stem cells (CSCs) are a subpopulation of cells in a tumor that can self-regenerate and produce different types of cells with the ability to initiate tumor growth and dissemination. Chemotherapy resistance, caused by numerous mechanisms by which tumor tissue manages to overcome the effects of drugs, remains the main problem in cancer treatment. The identification of markers on the cell surface specific to CSCs is important for understanding this phenomenon. (2) Methods: The expression of markers CD24, CD44, ALDH1, and ABCG2 was analyzed on the surface of CSCs in two cancer cell lines, MDA-MB-231 and HCT-116, after treatment with 5-fluorouracil (5-FU) using flow cytometry analysis. A machine learning model (ML)-genetic algorithm (GA) was used for the in silico simulation of drug resistance. (3) Results: As evaluated through the use of flow cytometry, the percentage of CD24-CD44+ MDA-MB-231 and CD44, ALDH1 and ABCG2 HCT-116 in a group treated with 5-FU was significantly increased compared to untreated cells. The CSC population was enriched after treatment with chemotherapy, suggesting that these cells have enhanced drug resistance mechanisms. (4) Conclusions: Each individual GA prediction model achieved high accuracy in estimating the expression rate of CSC markers on cancer cells treated with 5-FU. Artificial intelligence can be used as a powerful tool for predicting drug resistance.
Collapse
Affiliation(s)
- Amra Ramović Hamzagić
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Danijela Cvetković
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Marina Gazdić Janković
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Nevena Milivojević Dimitrijević
- Institute for Information Technologies Kragujevac, University of Kragujevac, Liceja Kneževine Srbije 1A, 34000 Kragujevac, Serbia; (N.M.D.); (D.N.); (M.Ž.); (D.Š.)
| | - Dalibor Nikolić
- Institute for Information Technologies Kragujevac, University of Kragujevac, Liceja Kneževine Srbije 1A, 34000 Kragujevac, Serbia; (N.M.D.); (D.N.); (M.Ž.); (D.Š.)
- Bioengineering Research and Development Center (BioIRC), Prvoslava Stojanovica 6, 34000 Kragujevac, Serbia;
| | - Marko Živanović
- Institute for Information Technologies Kragujevac, University of Kragujevac, Liceja Kneževine Srbije 1A, 34000 Kragujevac, Serbia; (N.M.D.); (D.N.); (M.Ž.); (D.Š.)
| | - Nikolina Kastratović
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ivica Petrović
- Faculty of Medical Sciences, Department of Pathophysiology, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Sandra Nikolić
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Milena Jovanović
- Faculty of Sciences, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia;
| | - Dragana Šeklić
- Institute for Information Technologies Kragujevac, University of Kragujevac, Liceja Kneževine Srbije 1A, 34000 Kragujevac, Serbia; (N.M.D.); (D.N.); (M.Ž.); (D.Š.)
| | - Nenad Filipović
- Bioengineering Research and Development Center (BioIRC), Prvoslava Stojanovica 6, 34000 Kragujevac, Serbia;
- Faculty of Engineering, University of Kragujevac, Sestre Janjić 6, 34000 Kragujevac, Serbia
| | - Biljana Ljujić
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
25
|
Liu M, Srivastava G, Ramanujam J, Brylinski M. SynerGNet: A Graph Neural Network Model to Predict Anticancer Drug Synergy. Biomolecules 2024; 14:253. [PMID: 38540674 PMCID: PMC10967862 DOI: 10.3390/biom14030253] [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: 12/24/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 01/03/2025] Open
Abstract
Drug combination therapy shows promise in cancer treatment by addressing drug resistance, reducing toxicity, and enhancing therapeutic efficacy. However, the intricate and dynamic nature of biological systems makes identifying potential synergistic drugs a costly and time-consuming endeavor. To facilitate the development of combination therapy, techniques employing artificial intelligence have emerged as a transformative solution, providing a sophisticated avenue for advancing existing therapeutic approaches. In this study, we developed SynerGNet, a graph neural network model designed to accurately predict the synergistic effect of drug pairs against cancer cell lines. SynerGNet utilizes cancer-specific featured graphs created by integrating heterogeneous biological features into the human protein-protein interaction network, followed by a reduction process to enhance topological diversity. Leveraging synergy data provided by AZ-DREAM Challenges, the model yields a balanced accuracy of 0.68, significantly outperforming traditional machine learning. Encouragingly, augmenting the training data with carefully constructed synthetic instances improved the balanced accuracy of SynerGNet to 0.73. Finally, the results of an independent validation conducted against DrugCombDB demonstrated that it exhibits a strong performance when applied to unseen data. SynerGNet shows a great potential in detecting drug synergy, positioning itself as a valuable tool that could contribute to the advancement of combination therapy for cancer treatment.
Collapse
Affiliation(s)
- Mengmeng Liu
- Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA; (M.L.)
| | - Gopal Srivastava
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - J. Ramanujam
- Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA; (M.L.)
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michal Brylinski
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
26
|
Bertão AR, Teixeira F, Ivasiv V, Parpot P, Almeida-Aguiar C, Fonseca AM, Bañobre-López M, Baltazar F, Neves IC. Machine Learning-Assisted Optimization of Drug Combinations in Zeolite-Based Delivery Systems for Melanoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:5696-5707. [PMID: 38271191 PMCID: PMC10859889 DOI: 10.1021/acsami.3c18224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/27/2024]
Abstract
Two independent artificial neural network (ANN) models were used to determine the optimal drug combination of zeolite-based delivery systems (ZDS) for cancer therapy. The systems were based on the NaY zeolite using silver (Ag+) and 5-fluorouracil (5-FU) as antimicrobial and antineoplastic agents. Different ZDS samples were prepared, and their characterization indicates the successful incorporation of both pharmacologically active species without any relevant changes to the zeolite structure. Silver acts as a counterion of the negative framework, and 5-FU retains its molecular integrity. The data from the A375 cell viability assays, involving ZDS samples (solid phase), 5-FU, and Ag+ aqueous solutions (liquid phase), were used to train two independent machine learning (ML) models. Both models exhibited a high level of accuracy in predicting the experimental cell viability results, allowing the development of a novel protocol for virtual cell viability assays. The findings suggest that the incorporation of both Ag and 5-FU into the zeolite structure significantly potentiates their anticancer activity when compared to that of the liquid phase. Additionally, two optimal AgY/5-FU@Y ratios were proposed to achieve the best cell viability outcomes. The ZDS also exhibited significant efficacy against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus); the predicted combination ratio is also effective against S. aureus, underscoring the potential of this approach as a therapeutic option for cancer-associated bacterial infections.
Collapse
Affiliation(s)
- Ana Raquel Bertão
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Life
and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s
- PT Government Associate Laboratory, University
of Minho, 4710-057 Braga/Guimarães, Portugal
- Advanced
(Magnetic) Theranostic Nanostructures Lab, Nanomedicine Group, International
Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| | - Filipe Teixeira
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Viktoriya Ivasiv
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Pier Parpot
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- CEB
- Centre of Biological Engineering, University
of Minho, 4710-057 Braga, Portugal
| | - Cristina Almeida-Aguiar
- CBMA - Centre
of Molecular and Environmental Biology, University of Minho, 4710-057 Braga, Portugal
| | - António M. Fonseca
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- CEB
- Centre of Biological Engineering, University
of Minho, 4710-057 Braga, Portugal
| | - Manuel Bañobre-López
- Advanced
(Magnetic) Theranostic Nanostructures Lab, Nanomedicine Group, International
Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| | - Fátima Baltazar
- Life
and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s
- PT Government Associate Laboratory, University
of Minho, 4710-057 Braga/Guimarães, Portugal
| | - Isabel C. Neves
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- CEB
- Centre of Biological Engineering, University
of Minho, 4710-057 Braga, Portugal
| |
Collapse
|
27
|
Galadima H, Anson-Dwamena R, Johnson A, Bello G, Adunlin G, Blando J. Machine Learning as a Tool for Early Detection: A Focus on Late-Stage Colorectal Cancer across Socioeconomic Spectrums. Cancers (Basel) 2024; 16:540. [PMID: 38339293 PMCID: PMC10854986 DOI: 10.3390/cancers16030540] [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: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
PURPOSE To assess the efficacy of various machine learning (ML) algorithms in predicting late-stage colorectal cancer (CRC) diagnoses against the backdrop of socio-economic and regional healthcare disparities. METHODS An innovative theoretical framework was developed to integrate individual- and census tract-level social determinants of health (SDOH) with sociodemographic factors. A comparative analysis of the ML models was conducted using key performance metrics such as AUC-ROC to evaluate their predictive accuracy. Spatio-temporal analysis was used to identify disparities in late-stage CRC diagnosis probabilities. RESULTS Gradient boosting emerged as the superior model, with the top predictors for late-stage CRC diagnosis being anatomic site, year of diagnosis, age, proximity to superfund sites, and primary payer. Spatio-temporal clusters highlighted geographic areas with a statistically significant high probability of late-stage diagnoses, emphasizing the need for targeted healthcare interventions. CONCLUSIONS This research underlines the potential of ML in enhancing the prognostic predictions in oncology, particularly in CRC. The gradient boosting model, with its robust performance, holds promise for deployment in healthcare systems to aid early detection and formulate localized cancer prevention strategies. The study's methodology demonstrates a significant step toward utilizing AI in public health to mitigate disparities and improve cancer care outcomes.
Collapse
Affiliation(s)
- Hadiza Galadima
- School of Community and Environmental Health, Old Dominion University, Norfolk, VA 23529, USA; (R.A.-D.); (A.J.); (J.B.)
| | - Rexford Anson-Dwamena
- School of Community and Environmental Health, Old Dominion University, Norfolk, VA 23529, USA; (R.A.-D.); (A.J.); (J.B.)
| | - Ashley Johnson
- School of Community and Environmental Health, Old Dominion University, Norfolk, VA 23529, USA; (R.A.-D.); (A.J.); (J.B.)
| | - Ghalib Bello
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Georges Adunlin
- Department of Pharmaceutical, Social and Administrative Sciences, Samford University, Birmingham, AL 35229, USA;
| | - James Blando
- School of Community and Environmental Health, Old Dominion University, Norfolk, VA 23529, USA; (R.A.-D.); (A.J.); (J.B.)
| |
Collapse
|
28
|
Liu M, Srivastava G, Ramanujam J, Brylinski M. Augmented drug combination dataset to improve the performance of machine learning models predicting synergistic anticancer effects. Sci Rep 2024; 14:1668. [PMID: 38238448 PMCID: PMC10796434 DOI: 10.1038/s41598-024-51940-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
Combination therapy has gained popularity in cancer treatment as it enhances the treatment efficacy and overcomes drug resistance. Although machine learning (ML) techniques have become an indispensable tool for discovering new drug combinations, the data on drug combination therapy currently available may be insufficient to build high-precision models. We developed a data augmentation protocol to unbiasedly scale up the existing anti-cancer drug synergy dataset. Using a new drug similarity metric, we augmented the synergy data by substituting a compound in a drug combination instance with another molecule that exhibits highly similar pharmacological effects. Using this protocol, we were able to upscale the AZ-DREAM Challenges dataset from 8798 to 6,016,697 drug combinations. Comprehensive performance evaluations show that ML models trained on the augmented data consistently achieve higher accuracy than those trained solely on the original dataset. Our data augmentation protocol provides a systematic and unbiased approach to generating more diverse and larger-scale drug combination datasets, enabling the development of more precise and effective ML models. The protocol presented in this study could serve as a foundation for future research aimed at discovering novel and effective drug combinations for cancer treatment.
Collapse
Affiliation(s)
- Mengmeng Liu
- Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Gopal Srivastava
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - J Ramanujam
- Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA, 70803, USA
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Michal Brylinski
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA.
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
29
|
Nasimian A, Younus S, Tatli Ö, Hammarlund EU, Pienta KJ, Rönnstrand L, Kazi JU. AlphaML: A clear, legible, explainable, transparent, and elucidative binary classification platform for tabular data. PATTERNS (NEW YORK, N.Y.) 2024; 5:100897. [PMID: 38264719 PMCID: PMC10801203 DOI: 10.1016/j.patter.2023.100897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/07/2023] [Accepted: 11/21/2023] [Indexed: 01/25/2024]
Abstract
Leveraging the potential of machine learning and recognizing the broad applications of binary classification, it becomes essential to develop platforms that are not only powerful but also transparent, interpretable, and user friendly. We introduce alphaML, a user-friendly platform that provides clear, legible, explainable, transparent, and elucidative (CLETE) binary classification models with comprehensive customization options. AlphaML offers feature selection, hyperparameter search, sampling, and normalization methods, along with 15 machine learning algorithms with global and local interpretation. We have integrated a custom metric for hyperparameter search that considers both training and validation scores, safeguarding against under- or overfitting. Additionally, we employ the NegLog2RMSL scoring method, which uses both training and test scores for a thorough model evaluation. The platform has been tested using datasets from multiple domains and offers a graphical interface, removing the need for programming expertise. Consequently, alphaML exhibits versatility, demonstrating promising applicability across a broad spectrum of tabular data configurations.
Collapse
Affiliation(s)
- Ahmad Nasimian
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| | - Saleena Younus
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| | - Özge Tatli
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| | - Emma U. Hammarlund
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
- Tissue Development and Evolution (TiDE), Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Kenneth J. Pienta
- The Cancer Ecology Center, Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lars Rönnstrand
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Julhash U. Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| |
Collapse
|
30
|
Dziubańska-Kusibab PJ, Nevedomskaya E, Haendler B. Preclinical Anticipation of On- and Off-Target Resistance Mechanisms to Anti-Cancer Drugs: A Systematic Review. Int J Mol Sci 2024; 25:705. [PMID: 38255778 PMCID: PMC10815614 DOI: 10.3390/ijms25020705] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/22/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
The advent of targeted therapies has led to tremendous improvements in treatment options and their outcomes in the field of oncology. Yet, many cancers outsmart precision drugs by developing on-target or off-target resistance mechanisms. Gaining the ability to resist treatment is the rule rather than the exception in tumors, and it remains a major healthcare challenge to achieve long-lasting remission in most cancer patients. Here, we discuss emerging strategies that take advantage of innovative high-throughput screening technologies to anticipate on- and off-target resistance mechanisms before they occur in treated cancer patients. We divide the methods into non-systematic approaches, such as random mutagenesis or long-term drug treatment, and systematic approaches, relying on the clustered regularly interspaced short palindromic repeats (CRISPR) system, saturated mutagenesis, or computational methods. All these new developments, especially genome-wide CRISPR-based screening platforms, have significantly accelerated the processes for identification of the mechanisms responsible for cancer drug resistance and opened up new avenues for future treatments.
Collapse
Affiliation(s)
| | | | - Bernard Haendler
- Research and Early Development Oncology, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany; (P.J.D.-K.); (E.N.)
| |
Collapse
|
31
|
Braga L, Lopes R, Alves L, Mota F. The global patent landscape of artificial intelligence applications for cancer. Nat Biotechnol 2023; 41:1679-1687. [PMID: 38082076 DOI: 10.1038/s41587-023-02051-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Affiliation(s)
- Luiza Braga
- Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Luiz Alves
- Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Fabio Mota
- Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
32
|
Zhou SN, Jv DW, Meng XF, Zhang JJ, Liu C, Wu ZY, Hong N, Lu YY, Zhang N. Feasibility of machine learning-based modeling and prediction using multiple centers data to assess intrahepatic cholangiocarcinoma outcomes. Ann Med 2023; 55:215-223. [PMID: 36576390 PMCID: PMC9809369 DOI: 10.1080/07853890.2022.2160008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND AIMS Currently, there are still no definitive consensus in the treatment of intrahepatic cholangiocarcinoma (iCCA). This study aimed to build a clinical decision support tool based on machine learning using the Surveillance, Epidemiology, and End Results (SEER) database and the data from the Fifth Medical Center of the PLA General Hospital in China. METHODS 4,398 eligible patients from the SEER database and 504 eligible patients from the hospital data, who presented with histologically proven iCCA, were enrolled for modeling by cross-validation based on machine learning. All the models were trained using the open-source Python library scikit-survival version 0.16.0. Shapley additive explanations method was used to help clinicians better understand the obtained results. Permutation importance was calculated using library ELI5. RESULTS All involved treatment modalities could contribute to a better prognosis. Three models were derived and tested using different data sources, with concordance indices of 0.67, 0.69, and 0.73, respectively. The prediction results were consistent with those under actual situations involving randomly selected patients. Model 2, trained using the hospital data, was selected to develop an online tool, due to its advantage in predicting short-term prognosis. CONCLUSION The prediction model and tool established in this study can be applied to predict the prognosis of iCCA after treatment by inputting the patient's clinical parameters or TNM stages and treatment options, thus contributing to optimal clinical decisions.KEY MESSAGESA prognostic model related to disease staging and treatment mode was conducted using the method of machine learning, based on the big data of multi centers.The online calculator can predict the short-term survival prognosis of intrahepatic cholangiocarcinoma, thus, help to make the best clinical decision.The online calculator built to calculate the mortality risk and overall survival can be easily obtained and applied.
Collapse
Affiliation(s)
- Shuang-Nan Zhou
- Senior Department of Infectious Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Da-Wei Jv
- The Fifth Out-patient Department, Central Theater Command General Hospital of Chinese PLA, Wuhan, Hubei, China
| | - Xiang-Fei Meng
- Faculty of Hepato-Pancrato-Biliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jing-Jing Zhang
- Senior Department of Liver Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chun Liu
- Digital Health China Technologies Co., Ltd, Beijing, China
| | - Ze-Yi Wu
- Digital Health China Technologies Co., Ltd, Beijing, China
| | - Na Hong
- Digital Health China Technologies Co., Ltd, Beijing, China
| | - Yin-Ying Lu
- Senior Department of Liver Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ning Zhang
- Senior Department of Liver Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
33
|
Al-Rajab M, Lu J, Xu Q, Kentour M, Sawsa A, Shuweikeh E, Joy M, Arasaradnam R. A hybrid machine learning feature selection model-HMLFSM to enhance gene classification applied to multiple colon cancers dataset. PLoS One 2023; 18:e0286791. [PMID: 37917732 PMCID: PMC10621932 DOI: 10.1371/journal.pone.0286791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/20/2023] [Indexed: 11/04/2023] Open
Abstract
Colon cancer is a significant global health problem, and early detection is critical for improving survival rates. Traditional detection methods, such as colonoscopies, can be invasive and uncomfortable for patients. Machine Learning (ML) algorithms have emerged as a promising approach for non-invasive colon cancer classification using genetic data or patient demographics and medical history. One approach is to use ML to analyse genetic data, or patient demographics and medical history, to predict the likelihood of colon cancer. However, due to the challenges imposed by variable gene expression and the high dimensionality of cancer-related datasets, traditional transductive ML applications have limited accuracy and risk overfitting. In this paper, we propose a new hybrid feature selection model called HMLFSM-Hybrid Machine Learning Feature Selection Model to improve colon cancer gene classification. We developed a multifilter hybrid model including a two-phase feature selection approach, combining Information Gain (IG) and Genetic Algorithms (GA), and minimum Redundancy Maximum Relevance (mRMR) coupling with Particle Swarm Optimization (PSO). We critically tested our model on three colon cancer genetic datasets and found that the new framework outperformed other models with significant accuracy improvements (95%, ~97%, and ~94% accuracies for datasets 1, 2, and 3 respectively). The results show that our approach improves the classification accuracy of colon cancer detection by highlighting important and relevant genes, eliminating irrelevant ones, and revealing the genes that have a direct influence on the classification process. For colon cancer gene analysis, and along with our experiments and literature review, we found that selective input feature extraction prior to feature selection is essential for improving predictive performance.
Collapse
Affiliation(s)
- Murad Al-Rajab
- College of Engineering, Abu Dhabi University, Abu Dhabi, United Arab Emirates
- School of Computing and Engineering, University of Huddersfield, Huddersfield, United Kingdom
| | - Joan Lu
- School of Computing and Engineering, University of Huddersfield, Huddersfield, United Kingdom
| | - Qiang Xu
- School of Computing and Engineering, University of Huddersfield, Huddersfield, United Kingdom
| | - Mohamed Kentour
- School of Computing and Engineering, University of Huddersfield, Huddersfield, United Kingdom
| | - Ahlam Sawsa
- School of Computing and Engineering, University of Huddersfield, Huddersfield, United Kingdom
- Bradford Teaching Hospitals NHS Foundation Trust, Bradford, United Kingdom
| | - Emad Shuweikeh
- School of Computing and Engineering, University of Huddersfield, Huddersfield, United Kingdom
| | - Mike Joy
- University of Warwick, Coventry, United Kingdom
| | | |
Collapse
|
34
|
Cui S, Wei G, Zhou L, Zhao E, Wang T, Ma F. Predicting line of therapy transition via similar patient augmentation. J Biomed Inform 2023; 147:104511. [PMID: 37813326 DOI: 10.1016/j.jbi.2023.104511] [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: 03/25/2023] [Revised: 07/28/2023] [Accepted: 09/29/2023] [Indexed: 10/11/2023]
Abstract
Analyzing large EHR databases to predict cancer progression and treatments has become a hot trend in recent years. An increasing number of modern deep learning models have been proposed to find the milestones of essential patient medical journey characteristics to predict their disease status and give healthcare professionals valuable insights. However, most of the existing methods are lack of consideration for the inter-relationship among different patients. We believe that more valuable information can be extracted, especially when patients with similar disease statuses visit the same doctors. Towards this end, a similar patient augmentation-based approach named SimPA is proposed to enhance the learning of patient representations and further predict lines of therapy transition. Our experiment results on a real-world multiple myeloma dataset show that our proposed approach outperforms state-of-the-art baseline approaches in terms of standard evaluation metrics for classification tasks.
Collapse
Affiliation(s)
- Suhan Cui
- College of Information Sciences and Technology, Pennsylvania State University, State College, PA, 16802, USA
| | - Guanhao Wei
- Advanced Analytics, IQVIA Inc, Wayne, PA, 19087, USA
| | - Li Zhou
- Advanced Analytics, IQVIA Inc, Wayne, PA, 19087, USA
| | - Emily Zhao
- Advanced Analytics, IQVIA Inc, Wayne, PA, 19087, USA
| | - Ting Wang
- College of Information Sciences and Technology, Pennsylvania State University, State College, PA, 16802, USA
| | - Fenglong Ma
- College of Information Sciences and Technology, Pennsylvania State University, State College, PA, 16802, USA.
| |
Collapse
|
35
|
Liu M, Srivastava G, Ramanujam J, Brylinski M. Augmented drug combination dataset to improve the performance of machine learning models predicting synergistic anticancer effects. RESEARCH SQUARE 2023:rs.3.rs-3481858. [PMID: 37961281 PMCID: PMC10635365 DOI: 10.21203/rs.3.rs-3481858/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Combination therapy has gained popularity in cancer treatment as it enhances the treatment efficacy and overcomes drug resistance. Although machine learning (ML) techniques have become an indispensable tool for discovering new drug combinations, the data on drug combination therapy currently available may be insufficient to build high-precision models. We developed a data augmentation protocol to unbiasedly scale up the existing anti-cancer drug synergy dataset. Using a new drug similarity metric, we augmented the synergy data by substituting a compound in a drug combination instance with another molecule that exhibits highly similar pharmacological effects. Using this protocol, we were able to upscale the AZ-DREAM Challenges dataset from 8,798 to 6,016,697 drug combinations. Comprehensive performance evaluations show that Random Forest and Gradient Boosting Trees models trained on the augmented data achieve higher accuracy than those trained solely on the original dataset. Our data augmentation protocol provides a systematic and unbiased approach to generating more diverse and larger-scale drug combination datasets, enabling the development of more precise and effective ML models. The protocol presented in this study could serve as a foundation for future research aimed at discovering novel and effective drug combinations for cancer treatment.
Collapse
|
36
|
Huang L, Ye X, Wu F, Wang X, Qiu M. Study of prevalence and risk factors of chemotherapy-induced mucositis in gastrointestinal cancer using machine learning models. Front Oncol 2023; 13:1138992. [PMID: 37841443 PMCID: PMC10569816 DOI: 10.3389/fonc.2023.1138992] [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: 01/06/2023] [Accepted: 08/30/2023] [Indexed: 10/17/2023] Open
Abstract
Objective Chemotherapy-induced mucositis (CIM) significantly impacts clinical outcomes and diminishes the quality of life in patients with gastrointestinal cancer. This study aims to prospectively determine the incidence, severity, and underlying risk factors associated with CIM in this patient population. Methods To achieve this objective, we introduce a novel Machine Learning-based Toxicity Prediction Model (ML-TPM) designed to analyze the risk factors contributing to CIM development in gastrointestinal cancer patients. Within the winter season spanning from December 15th, 2018 to January 14th, 2019, we conducted in-person interviews with patients undergoing chemotherapy for gastrointestinal cancer. These interviews encompassed comprehensive questionnaires pertaining to patient demographics, CIM incidence, severity, and any supplementary prophylactic measures employed. Results The study encompassed a cohort of 447 participating patients who provided complete questionnaire responses (100%). Of these, 328 patients (73.4%) reported experiencing CIM during the course of their treatment. Notably, CIM-induced complications led to treatment discontinuation in 14 patients (3%). The most frequently encountered CIM symptoms were diarrhea (41.6%), followed by nausea (37.8%), vomiting (25.1%), abdominal pain (21%), gastritis (10.5%), and oral pain (10.3%). Supplementary prophylaxis was administered to approximately 62% of the patients. The analysis revealed significant correlations between the overall incidence of CIM and gender (p=0.015), number of chemotherapy cycles exceeding one (p=0.039), utilization of platinum-based regimens (p=0.039), and administration of irinotecan (p=0.003). Specifically, the incidence of diarrhea exhibited positive correlations with prior surgical history (p=0.037), irinotecan treatment (p=0.021), and probiotics usage (p=0.035). Conversely, diarrhea incidence demonstrated an adverse correlation with platinum-based treatment (p=0.026). Conclusion In conclusion, this study demonstrates the successful implementation of the ML-TPM model for automating toxicity prediction with accuracy comparable to conventional physical analyses. Our findings provide valuable insights into the identification of CIM risk factors among gastrointestinal cancer patients undergoing chemotherapy. Furthermore, the results underscore the potential of machine learning in enhancing our understanding of chemotherapy-induced mucositis and advancing personalized patient care strategies.
Collapse
Affiliation(s)
- Lin Huang
- Division of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xianhui Ye
- Division of Medical Oncology, Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Fengqing Wu
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiuyun Wang
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Meng Qiu
- Division of Medical Oncology, Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
37
|
Shah K, Nasimian A, Ahmed M, Al Ashiri L, Denison L, Sime W, Bendak K, Kolosenko I, Siino V, Levander F, Palm-Apergi C, Massoumi R, Lock RB, Kazi JU. PLK1 as a cooperating partner for BCL2-mediated antiapoptotic program in leukemia. Blood Cancer J 2023; 13:139. [PMID: 37679323 PMCID: PMC10484999 DOI: 10.1038/s41408-023-00914-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/15/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
The deregulation of BCL2 family proteins plays a crucial role in leukemia development. Therefore, pharmacological inhibition of this family of proteins is becoming a prevalent treatment method. However, due to the emergence of primary and acquired resistance, efficacy is compromised in clinical or preclinical settings. We developed a drug sensitivity prediction model utilizing a deep tabular learning algorithm for the assessment of venetoclax sensitivity in T-cell acute lymphoblastic leukemia (T-ALL) patient samples. Through analysis of predicted venetoclax-sensitive and resistant samples, PLK1 was identified as a cooperating partner for the BCL2-mediated antiapoptotic program. This finding was substantiated by additional data obtained through phosphoproteomics and high-throughput kinase screening. Concurrent treatment using venetoclax with PLK1-specific inhibitors and PLK1 knockdown demonstrated a greater therapeutic effect on T-ALL cell lines, patient-derived xenografts, and engrafted mice compared with using each treatment separately. Mechanistically, the attenuation of PLK1 enhanced BCL2 inhibitor sensitivity through upregulation of BCL2L13 and PMAIP1 expression. Collectively, these findings underscore the dependency of T-ALL on PLK1 and postulate a plausible regulatory mechanism.
Collapse
Affiliation(s)
- Kinjal Shah
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ahmad Nasimian
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Mehreen Ahmed
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Lina Al Ashiri
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Linn Denison
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Wondossen Sime
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Katerina Bendak
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Iryna Kolosenko
- Department of Laboratory Medicine, Biomolecular & Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Valentina Siino
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Fredrik Levander
- Department of Immunotechnology, Lund University, Lund, Sweden
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Lund University, Lund, Sweden
| | - Caroline Palm-Apergi
- Department of Laboratory Medicine, Biomolecular & Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ramin Massoumi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Julhash U Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden.
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
38
|
Flanary VL, Fisher JL, Wilk EJ, Howton TC, Lasseigne BN. Computational Advancements in Cancer Combination Therapy Prediction. JCO Precis Oncol 2023; 7:e2300261. [PMID: 37824797 DOI: 10.1200/po.23.00261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/20/2023] [Accepted: 08/15/2023] [Indexed: 10/14/2023] Open
Abstract
Given the high attrition rate of de novo drug discovery and limited efficacy of single-agent therapies in cancer treatment, combination therapy prediction through in silico drug repurposing has risen as a time- and cost-effective alternative for identifying novel and potentially efficacious therapies for cancer. The purpose of this review is to provide an introduction to computational methods for cancer combination therapy prediction and to summarize recent studies that implement each of these methods. A systematic search of the PubMed database was performed, focusing on studies published within the past 10 years. Our search included reviews and articles of ongoing and retrospective studies. We prioritized articles with findings that suggest considerations for improving combination therapy prediction methods over providing a meta-analysis of all currently available cancer combination therapy prediction methods. Computational methods used for drug combination therapy prediction in cancer research include networks, regression-based machine learning, classifier machine learning models, and deep learning approaches. Each method class has its own advantages and disadvantages, so careful consideration is needed to determine the most suitable class when designing a combination therapy prediction method. Future directions to improve current combination therapy prediction technology include incorporation of disease pathobiology, drug characteristics, patient multiomics data, and drug-drug interactions to determine maximally efficacious and tolerable drug regimens for cancer. As computational methods improve in their capability to integrate patient, drug, and disease data, more comprehensive models can be developed to more accurately predict safe and efficacious combination drug therapies for cancer and other complex diseases.
Collapse
Affiliation(s)
- Victoria L Flanary
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Jennifer L Fisher
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Elizabeth J Wilk
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Timothy C Howton
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Brittany N Lasseigne
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
39
|
Shahab M, Zheng G, Khan A, Wei D, Novikov AS. Machine Learning-Based Virtual Screening and Molecular Simulation Approaches Identified Novel Potential Inhibitors for Cancer Therapy. Biomedicines 2023; 11:2251. [PMID: 37626747 PMCID: PMC10452548 DOI: 10.3390/biomedicines11082251] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Cyclin-dependent kinase 2 (CDK2) is a promising target for cancer treatment, developing new effective CDK2 inhibitors is of great significance in anticancer therapy. The involvement of CDK2 in tumorigenesis has been debated, but recent evidence suggests that specifically inhibiting CDK2 could be beneficial in treating certain tumors. This approach remains attractive in the development of anticancer drugs. Several small-molecule inhibitors targeting CDK2 have reached clinical trials, but a selective inhibitor for CDK2 is yet to be discovered. In this study, we conducted machine learning-based drug designing to search for a drug candidate for CDK2. Machine learning models, including k-NN, SVM, RF, and GNB, were created to detect active and inactive inhibitors for a CDK2 drug target. The models were assessed using 10-fold cross-validation to ensure their accuracy and reliability. These methods are highly suitable for classifying compounds as either active or inactive through the virtual screening of extensive compound libraries. Subsequently, machine learning techniques were employed to analyze the test dataset obtained from the zinc database. A total of 25 compounds with 98% accuracy were predicted as active against CDK2. These compounds were docked into CDK2's active site. Finally, three compounds were selected based on good docking score, and, along with a reference compound, underwent MD simulation. The Gaussian naïve Bayes model yielded superior results compared to other models. The top three hits exhibited enhanced stability and compactness compared to the reference compound. In conclusion, our study provides valuable insights for identifying and refining lead compounds as CDK2 inhibitors.
Collapse
Affiliation(s)
- Muhammad Shahab
- State Key Laboratories of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing 100029, China;
| | - Guojun Zheng
- State Key Laboratories of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing 100029, China;
| | - Abbas Khan
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; (A.K.); (D.W.)
| | - Dongqing Wei
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; (A.K.); (D.W.)
| | - Alexander S. Novikov
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia
- Research Institute of Chemistry, Peoples’ Friendship University of Russia (RUDN University), Moscow 117198, Russia
| |
Collapse
|
40
|
Sun B, Chen L. Interpretable deep learning for improving cancer patient survival based on personal transcriptomes. Sci Rep 2023; 13:11344. [PMID: 37443344 PMCID: PMC10344908 DOI: 10.1038/s41598-023-38429-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 07/07/2023] [Indexed: 07/15/2023] Open
Abstract
Precision medicine chooses the optimal drug for a patient by considering individual differences. With the tremendous amount of data accumulated for cancers, we develop an interpretable neural network to predict cancer patient survival based on drug prescriptions and personal transcriptomes (CancerIDP). The deep learning model achieves 96% classification accuracy in distinguishing short-lived from long-lived patients. The Pearson correlation between predicted and actual months-to-death values is as high as 0.937. About 27.4% of patients may survive longer with an alternative medicine chosen by our deep learning model. The median survival time of all patients can increase by 3.9 months. Our interpretable neural network model reveals the most discriminating pathways in the decision-making process, which will further facilitate mechanistic studies of drug development for cancers.
Collapse
Affiliation(s)
- Bo Sun
- Department of Quantitative and Computational Biology, University of Southern California, 1050 Childs Way, Los Angeles, CA, 90089, USA
| | - Liang Chen
- Department of Quantitative and Computational Biology, University of Southern California, 1050 Childs Way, Los Angeles, CA, 90089, USA.
| |
Collapse
|
41
|
Si H, Esquivel M, Mendoza Mendoza E, Roarty K. The covert symphony: cellular and molecular accomplices in breast cancer metastasis. Front Cell Dev Biol 2023; 11:1221784. [PMID: 37440925 PMCID: PMC10333702 DOI: 10.3389/fcell.2023.1221784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer has emerged as the most commonly diagnosed cancer and primary cause of cancer-related deaths among women worldwide. Although significant progress has been made in targeting the primary tumor, the effectiveness of systemic treatments to prevent metastasis remains limited. Metastatic disease continues to be the predominant factor leading to fatality in the majority of breast cancer patients. The existence of a prolonged latency period between initial treatment and eventual recurrence in certain patients indicates that tumors can both adapt to and interact with the systemic environment of the host, facilitating and sustaining the progression of the disease. In order to identify potential therapeutic interventions for metastasis, it will be crucial to gain a comprehensive framework surrounding the mechanisms driving the growth, survival, and spread of tumor cells, as well as their interaction with supporting cells of the microenvironment. This review aims to consolidate recent discoveries concerning critical aspects of breast cancer metastasis, encompassing the intricate network of cells, molecules, and physical factors that contribute to metastasis, as well as the molecular mechanisms governing cancer dormancy.
Collapse
Affiliation(s)
- Hongjiang Si
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Madelyn Esquivel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Erika Mendoza Mendoza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Kevin Roarty
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| |
Collapse
|
42
|
Liu J, Wu P, Lai S, Wang J, Hou H, Zhang Y. Prognostic models for upper urinary tract urothelial carcinoma patients after radical nephroureterectomy based on a novel systemic immune-inflammation score with machine learning. BMC Cancer 2023; 23:574. [PMID: 37349696 DOI: 10.1186/s12885-023-11058-z] [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: 11/19/2022] [Accepted: 06/11/2023] [Indexed: 06/24/2023] Open
Abstract
PURPOSE This study aimed to evaluate the clinical significance of a novel systemic immune-inflammation score (SIIS) to predict oncological outcomes in upper urinary tract urothelial carcinoma(UTUC) after radical nephroureterectomy(RNU). METHOD The clinical data of 483 patients with nonmetastatic UTUC underwent surgery in our center were analyzed. Five inflammation-related biomarkers were screened in the Lasso-Cox model and then aggregated to generate the SIIS based on the regression coefficients. Overall survival (OS) was assessed using Kaplan-Meier analyses. The Cox proportional hazards regression and random survival forest model were adopted to build the prognostic model. Then we established an effective nomogram for UTUC after RNU based on SIIS. The discrimination and calibration of the nomogram were evaluated using the concordance index (C-index), area under the time-dependent receiver operating characteristic curve (time-dependent AUC), and calibration curves. Decision curve analysis (DCA) was used to assess the net benefits of the nomogram at different threshold probabilities. RESULT According to the median value SIIS computed by the lasso Cox model, the high-risk group had worse OS (p<0.0001) than low risk-group. Variables with a minimum depth greater than the depth threshold or negative variable importance were excluded, and the remaining six variables were included in the model. The area under the ROC curve (AUROC) of the Cox and random survival forest models were 0.801 and 0.872 for OS at five years, respectively. Multivariate Cox analysis showed that elevated SIIS was significantly associated with poorer OS (p<0.001). In terms of predicting overall survival, a nomogram that considered the SIIS and clinical prognostic factors performed better than the AJCC staging. CONCLUSION The pretreatment levels of SIIS were an independent predictor of prognosis in upper urinary tract urothelial carcinoma after RNU. Therefore, incorporating SIIS into currently available clinical parameters helps predict the long-term survival of UTUC.
Collapse
Affiliation(s)
- Jianyong Liu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Hospital Continence Center, Beijing, China
| | - Pengjie Wu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Hospital Continence Center, Beijing, China
| | - Shicong Lai
- Department of Urology, Peking University People's Hospital, 100044, Beijing, China
| | - Jianye Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, China.
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
- Beijing Hospital Continence Center, Beijing, China.
| | - Huimin Hou
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, China.
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
- Beijing Hospital Continence Center, Beijing, China.
| | - Yaoguang Zhang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, China.
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
- Beijing Hospital Continence Center, Beijing, China.
| |
Collapse
|
43
|
Singh AV, Chandrasekar V, Paudel N, Laux P, Luch A, Gemmati D, Tissato V, Prabhu KS, Uddin S, Dakua SP. Integrative toxicogenomics: Advancing precision medicine and toxicology through artificial intelligence and OMICs technology. Biomed Pharmacother 2023; 163:114784. [PMID: 37121152 DOI: 10.1016/j.biopha.2023.114784] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/15/2023] [Accepted: 04/24/2023] [Indexed: 05/02/2023] Open
Abstract
More information about a person's genetic makeup, drug response, multi-omics response, and genomic response is now available leading to a gradual shift towards personalized treatment. Additionally, the promotion of non-animal testing has fueled the computational toxicogenomics as a pivotal part of the next-gen risk assessment paradigm. Artificial Intelligence (AI) has the potential to provid new ways analyzing the patient data and making predictions about treatment outcomes or toxicity. As personalized medicine and toxicogenomics involve huge data processing, AI can expedite this process by providing powerful data processing, analysis, and interpretation algorithms. AI can process and integrate a multitude of data including genome data, patient records, clinical data and identify patterns to derive predictive models anticipating clinical outcomes and assessing the risk of any personalized medicine approaches. In this article, we have studied the current trends and future perspectives in personalized medicine & toxicology, the role of toxicogenomics in connecting the two fields, and the impact of AI on personalized medicine & toxicology. In this work, we also study the key challenges and limitations in personalized medicine, toxicogenomics, and AI in order to fully realize their potential.
Collapse
Affiliation(s)
- Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | | | - Namuna Paudel
- Department of Chemistry, Amrit Campus, Institute of Science and Technology, Tribhuvan University, Lainchaur, Kathmandu 44600 Nepal
| | - Peter Laux
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Andreas Luch
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Donato Gemmati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; Centre Hemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy; Centre for Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Veronica Tissato
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; Centre Hemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy; Centre for Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | |
Collapse
|
44
|
Verhaegen F, Butterworth KT, Chalmers AJ, Coppes RP, de Ruysscher D, Dobiasch S, Fenwick JD, Granton PV, Heijmans SHJ, Hill MA, Koumenis C, Lauber K, Marples B, Parodi K, Persoon LCGG, Staut N, Subiel A, Vaes RDW, van Hoof S, Verginadis IL, Wilkens JJ, Williams KJ, Wilson GD, Dubois LJ. Roadmap for precision preclinical x-ray radiation studies. Phys Med Biol 2023; 68:06RM01. [PMID: 36584393 DOI: 10.1088/1361-6560/acaf45] [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: 05/12/2022] [Accepted: 12/30/2022] [Indexed: 12/31/2022]
Abstract
This Roadmap paper covers the field of precision preclinical x-ray radiation studies in animal models. It is mostly focused on models for cancer and normal tissue response to radiation, but also discusses other disease models. The recent technological evolution in imaging, irradiation, dosimetry and monitoring that have empowered these kinds of studies is discussed, and many developments in the near future are outlined. Finally, clinical translation and reverse translation are discussed.
Collapse
Affiliation(s)
- Frank Verhaegen
- MAASTRO Clinic, Radiotherapy Division, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
- SmART Scientific Solutions BV, Maastricht, The Netherlands
| | - Karl T Butterworth
- Patrick G. Johnston, Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Anthony J Chalmers
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Rob P Coppes
- Departments of Biomedical Sciences of Cells & Systems, Section Molecular Cell Biology and Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Dirk de Ruysscher
- MAASTRO Clinic, Radiotherapy Division, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sophie Dobiasch
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine and Klinikum rechts der Isar, Germany
- Department of Medical Physics, Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Germany
| | - John D Fenwick
- Department of Medical Physics & Biomedical Engineering University College LondonMalet Place Engineering Building, London WC1E 6BT, United Kingdom
| | | | | | - Mark A Hill
- MRC Oxford Institute for Radiation Oncology, University of Oxford, ORCRB Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU München, Munich, Germany
- German Cancer Consortium (DKTK), Partner site Munich, Germany
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester, NY, United States of America
| | - Katia Parodi
- German Cancer Consortium (DKTK), Partner site Munich, Germany
- Department of Medical Physics, Faculty of Physics, Ludwig-Maximilians-Universität München, Garching b. Munich, Germany
| | | | - Nick Staut
- SmART Scientific Solutions BV, Maastricht, The Netherlands
| | - Anna Subiel
- National Physical Laboratory, Medical Radiation Science Hampton Road, Teddington, Middlesex, TW11 0LW, United Kingdom
| | - Rianne D W Vaes
- MAASTRO Clinic, Radiotherapy Division, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | - Ioannis L Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jan J Wilkens
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine and Klinikum rechts der Isar, Germany
- Physics Department, Technical University of Munich (TUM), Germany
| | - Kaye J Williams
- Division of Pharmacy and Optometry, University of Manchester, Manchester, United Kingdom
| | - George D Wilson
- Department of Radiation Oncology, Beaumont Health, MI, United States of America
- Henry Ford Health, Detroit, MI, United States of America
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
45
|
Partin A, Brettin TS, Zhu Y, Narykov O, Clyde A, Overbeek J, Stevens RL. Deep learning methods for drug response prediction in cancer: Predominant and emerging trends. Front Med (Lausanne) 2023; 10:1086097. [PMID: 36873878 PMCID: PMC9975164 DOI: 10.3389/fmed.2023.1086097] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Cancer claims millions of lives yearly worldwide. While many therapies have been made available in recent years, by in large cancer remains unsolved. Exploiting computational predictive models to study and treat cancer holds great promise in improving drug development and personalized design of treatment plans, ultimately suppressing tumors, alleviating suffering, and prolonging lives of patients. A wave of recent papers demonstrates promising results in predicting cancer response to drug treatments while utilizing deep learning methods. These papers investigate diverse data representations, neural network architectures, learning methodologies, and evaluations schemes. However, deciphering promising predominant and emerging trends is difficult due to the variety of explored methods and lack of standardized framework for comparing drug response prediction models. To obtain a comprehensive landscape of deep learning methods, we conducted an extensive search and analysis of deep learning models that predict the response to single drug treatments. A total of 61 deep learning-based models have been curated, and summary plots were generated. Based on the analysis, observable patterns and prevalence of methods have been revealed. This review allows to better understand the current state of the field and identify major challenges and promising solution paths.
Collapse
Affiliation(s)
- Alexander Partin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Thomas S. Brettin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Yitan Zhu
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Oleksandr Narykov
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Austin Clyde
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Jamie Overbeek
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Rick L. Stevens
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
- Department of Computer Science, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
46
|
Nasimian A, Al Ashiri L, Ahmed M, Duan H, Zhang X, Rönnstrand L, Kazi JU. A Receptor Tyrosine Kinase Inhibitor Sensitivity Prediction Model Identifies AXL Dependency in Leukemia. Int J Mol Sci 2023; 24:ijms24043830. [PMID: 36835239 PMCID: PMC9959897 DOI: 10.3390/ijms24043830] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/05/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
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 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.
Collapse
Affiliation(s)
- Ahmad Nasimian
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
| | - Lina Al Ashiri
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
| | - Mehreen Ahmed
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
| | - Hongzhi Duan
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
| | - Xiaoyue Zhang
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
| | - Lars Rönnstrand
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, 22185 Lund, Sweden
| | - Julhash U. Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
- Correspondence: ; Tel.: +46-462226407
| |
Collapse
|
47
|
Choudhary R, Walhekar V, Muthal A, Kumar D, Bagul C, Kulkarni R. Machine learning facilitated structural activity relationship approach for the discovery of novel inhibitors targeting EGFR. J Biomol Struct Dyn 2023; 41:12445-12463. [PMID: 36762704 DOI: 10.1080/07391102.2023.2175263] [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: 11/10/2022] [Accepted: 01/03/2023] [Indexed: 02/11/2023]
Abstract
This research manuscript aims to find the most effective epidermal growth factor receptor (EGFR) inhibitors from millions of in house compounds through Machine Learning (ML) techniques. ML-based structure activity relationship (SAR) models were validated to predict biological activity of untested novel molecules. Six ML algorithms, including k nearest neighbour (KNN), decision tree (DT), Logistic Regression, support vector machine (SVM), multilinear regression (MLR), and random forest (RF), were used to build for activity prediction. Among these, RF classifier (accuracy for train and test set is 90% and 81%) and RF regressor (R2 and MSE for trainset is 0.83 and 0.29 and for test set, 0.69 and 0.46) showed good predictive performance. Also, the six most essential features that affect the biological activity parameter and highly contribute to model development were successfully selected by the variable importance technique. RF regression model was used to predict the biological activity expressed as pIC50 of nearly ten million molecules while RF classification model classifies those molecules into active, moderately active, and least active according to their predicted pIC50. Based on two models, thousand molecules from million molecules with higher predicted pIC50 values and classified as active were selected for molecular docking. Based on the docking scores, predicted pIC50, and binding interactions with MET769 residue, compounds, i.e., Zinc257233137, Zinc257232249, and Zinc101379788, were identified as potential EGFR inhibitors with predicted pIC50 7.72, 7.85, and 7.70. Dynamics studies were also performed on Zinc257233137 to illustrate that it has good binding free energy and stable hydrogen bonding interactions with EGFR. These molecules can be used for further research and proved to be the novel drugs for EGFR in cancer treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rekha Choudhary
- Department of Pharmaceutical Chemistry, BVDU'S Poona College of Pharmacy, Pune, Maharashtra, India
| | - Vinayak Walhekar
- Department of Pharmaceutical Chemistry, BVDU'S Poona College of Pharmacy, Pune, Maharashtra, India
| | - Amol Muthal
- Department of Pharmacology, BVDU'S Poona College of Pharmacy, Pune, Maharashtra, India
| | - Dilip Kumar
- Department of Pharmaceutical Chemistry, BVDU'S Poona College of Pharmacy, Pune, Maharashtra, India
- Department of Entomology, University of California, Davis, Davis, California, USA
- UC Davis Comprehensive Cancer Centre, University of California, Davis, Davis, California, USA
| | - Chandrakant Bagul
- Department of Pharmaceutical Chemistry, BVDU'S Poona College of Pharmacy, Pune, Maharashtra, India
| | - Ravindra Kulkarni
- Department of Pharmaceutical Chemistry, BVDU'S Poona College of Pharmacy, Pune, Maharashtra, India
| |
Collapse
|
48
|
Nasimian A, Ahmed M, Hedenfalk I, Kazi JU. A deep tabular data learning model predicting cisplatin sensitivity identifies BCL2L1 dependency in cancer. Comput Struct Biotechnol J 2023; 21:956-964. [PMID: 36733702 PMCID: PMC9876747 DOI: 10.1016/j.csbj.2023.01.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/15/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023] Open
Abstract
Cisplatin, a platinum-based chemotherapeutic agent, is widely used as a front-line treatment for several malignancies. However, treatment outcomes vary widely due to intrinsic and acquired resistance. In this study, cisplatin-perturbed gene expression and pathway enrichment were used to define a gene signature, which was further utilized to develop a cisplatin sensitivity prediction model using the TabNet algorithm. The TabNet model performed better (>80 % accuracy) than all other machine learning models when compared to a wide range of machine learning algorithms. Moreover, by using feature importance and comparing predicted ovarian cancer patient samples, BCL2L1 was identified as an important gene contributing to cisplatin resistance. Furthermore, the pharmacological inhibition of BCL2L1 was found to synergistically increase cisplatin efficacy. Collectively, this study developed a tool to predict cisplatin sensitivity using cisplatin-perturbed gene expression and pathway enrichment knowledge and identified BCL2L1 as an important gene in this setting.
Collapse
Affiliation(s)
- Ahmad Nasimian
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden,Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Mehreen Ahmed
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden,Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, 223 81 Lund, Sweden
| | - Julhash U. Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden,Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden,Correspondence to: Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon village Building 404:C3, Scheelevägen 8, 22363 Lund, Sweden.
| |
Collapse
|
49
|
Classification and diagnostic prediction of breast cancer metastasis on clinical data using machine learning algorithms. Sci Rep 2023; 13:485. [PMID: 36627367 PMCID: PMC9831019 DOI: 10.1038/s41598-023-27548-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Metastatic Breast Cancer (MBC) is one of the primary causes of cancer-related deaths in women. Despite several limitations, histopathological information about the malignancy is used for the classification of cancer. The objective of our study is to develop a non-invasive breast cancer classification system for the diagnosis of cancer metastases. The anaconda-Jupyter notebook is used to develop various python programming modules for text mining, data processing, and Machine Learning (ML) methods. Utilizing classification model cross-validation criteria, including accuracy, AUC, and ROC, the prediction performance of the ML models is assessed. Welch Unpaired t-test was used to ascertain the statistical significance of the datasets. Text mining framework from the Electronic Medical Records (EMR) made it easier to separate the blood profile data and identify MBC patients. Monocytes revealed a noticeable mean difference between MBC patients as compared to healthy individuals. The accuracy of ML models was dramatically improved by removing outliers from the blood profile data. A Decision Tree (DT) classifier displayed an accuracy of 83% with an AUC of 0.87. Next, we deployed DT classifiers using Flask to create a web application for robust diagnosis of MBC patients. Taken together, we conclude that ML models based on blood profile data may assist physicians in selecting intensive-care MBC patients to enhance the overall survival outcome.
Collapse
|
50
|
Xie R, Liu L, Lu X, He C, Li G. Identification of the diagnostic genes and immune cell infiltration characteristics of gastric cancer using bioinformatics analysis and machine learning. Front Genet 2023; 13:1067524. [PMID: 36685898 PMCID: PMC9845288 DOI: 10.3389/fgene.2022.1067524] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Background: Finding reliable diagnostic markers for gastric cancer (GC) is important. This work uses machine learning (ML) to identify GC diagnostic genes and investigate their connection with immune cell infiltration. Methods: We downloaded eight GC-related datasets from GEO, TCGA, and GTEx. GSE13911, GSE15459, GSE19826, GSE54129, and GSE79973 were used as the training set, GSE66229 as the validation set A, and TCGA & GTEx as the validation set B. First, the training set screened differentially expressed genes (DEGs), and gene ontology (GO), kyoto encyclopedia of genes and genomes (KEGG), disease Ontology (DO), and gene set enrichment analysis (GSEA) analyses were performed. Then, the candidate diagnostic genes were screened by LASSO and SVM-RFE algorithms, and receiver operating characteristic (ROC) curves evaluated the diagnostic efficacy. Then, the infiltration characteristics of immune cells in GC samples were analyzed by CIBERSORT, and correlation analysis was performed. Finally, mutation and survival analyses were performed for diagnostic genes. Results: We found 207 up-regulated genes and 349 down-regulated genes among 556 DEGs. gene ontology analysis significantly enriched 413 functional annotations, including 310 biological processes, 23 cellular components, and 80 molecular functions. Six of these biological processes are closely related to immunity. KEGG analysis significantly enriched 11 signaling pathways. 244 diseases were closely related to Ontology analysis. Multiple entries of the gene set enrichment analysis analysis were closely related to immunity. Machine learning screened eight candidate diagnostic genes and further validated them to identify ABCA8, COL4A1, FAP, LY6E, MAMDC2, and TMEM100 as diagnostic genes. Six diagnostic genes were mutated to some extent in GC. ABCA8, COL4A1, LY6E, MAMDC2, TMEM100 had prognostic value. Conclusion: We screened six diagnostic genes for gastric cancer through bioinformatic analysis and machine learning, which are intimately related to immune cell infiltration and have a definite prognostic value.
Collapse
Affiliation(s)
- Rongjun Xie
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China,Department of General Surgery, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China,Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Longfei Liu
- Department of General Surgery, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xianzhou Lu
- Department of General Surgery, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Chengjian He
- Department of Intensive Care Medicine, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China,*Correspondence: Guoxin Li,
| |
Collapse
|