1
|
Díaz-Santiago E, Moya-García AA, Pérez-García J, Yahyaoui R, Orengo C, Pazos F, Perkins JR, Ranea JAG. Better understanding the phenotypic effects of drugs through shared targets in genetic disease networks. Front Pharmacol 2025; 15:1470931. [PMID: 39911831 PMCID: PMC11794328 DOI: 10.3389/fphar.2024.1470931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/12/2024] [Indexed: 02/07/2025] Open
Abstract
Introduction Most drugs fail during development and there is a clear and unmet need for approaches to better understand mechanistically how drugs exert both their intended and adverse effects. Gaining traction in this field is the use of disease data linking genes with pathological phenotypes and combining this with drugtarget interaction data. Methods We introduce methodology to associate drugs with effects, both intended and adverse, using a tripartite network approach that combines drug-target and target-phenotype data, in which targets can be represented as proteins and protein domains. Results We were able to detect associations for over 140,000 ChEMBL drugs and 3,800 phenotypes, represented as Human Phenotype Ontology (HPO) terms. The overlap of these results with the SIDER databases of known drug side effects was up to 10 times higher than random, depending on the target type, disease database and score threshold used. In terms of overlap with drug-phenotype pairs extracted from the literature, the performance of our methodology was up to 17.47 times greater than random. The top results include phenotype-drug associations that represent intended effects, particularly for cancers such as chronic myelogenous leukemia, which was linked with nilotinib. They also include adverse side effects, such as blurred vision being linked with tetracaine. Discussion This work represents an important advance in our understanding of how drugs cause intended and adverse side effects through their action on disease causing genes and has potential applications for drug development and repositioning.
Collapse
Affiliation(s)
- Elena Díaz-Santiago
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
| | | | - Jesús Pérez-García
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
| | - Raquel Yahyaoui
- Laboratory of Inherited Metabolic Diseases and Newborn Screening, Malaga Regional University Hospital, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
| | - Christine Orengo
- Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Florencio Pazos
- Computational Systems Biology Group, Systems Biology Department, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| | - James R. Perkins
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan A. G. Ranea
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Spanish National Bioinformatics Institute (INB/ELIXIR-ES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
2
|
Halu A, Chelvanambi S, Decano JL, Matamalas JT, Whelan M, Asano T, Kalicharran N, Singh SA, Loscalzo J, Aikawa M. Integrating pharmacogenomics and cheminformatics with diverse disease phenotypes for cell type-guided drug discovery. Genome Med 2025; 17:7. [PMID: 39833831 PMCID: PMC11744892 DOI: 10.1186/s13073-025-01431-x] [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: 01/19/2023] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Large-scale pharmacogenomic resources, such as the Connectivity Map (CMap), have greatly assisted computational drug discovery. However, despite their widespread use, CMap-based methods have thus far been agnostic to the biological activity of drugs as well as to the genomic effects of drugs in multiple disease contexts. Here, we present a network-based statistical approach, Pathopticon, that uses CMap to build cell type-specific gene-drug perturbation networks and integrates these networks with cheminformatic data and diverse disease phenotypes to prioritize drugs in a cell type-dependent manner. METHODS We build cell type-specific gene-drug perturbation networks from CMap data using a statistical procedure we call Quantile-based Instance Z-score Consensus (QUIZ-C). Using these networks and a large-scale disease-gene network consisting of 569 disease signatures from the Enrichr database, we calculate Pathophenotypic Congruity Scores (PACOS) between input gene signatures and drug perturbation signatures and combine these scores with cheminformatic data from ChEMBL to prioritize drugs. We benchmark our approach by calculating area under the receiver operating characteristic curves (AUROC) for 73 gene sets from the Molecular Signatures Database (MSigDB) using target gene expression profiles from the Comparative Toxicogenomics Database (CTD). We validate the drugs predicted in our proofs-of-concept using real-time polymerase chain reaction (qPCR) experiments. RESULTS Cell type-specific gene-drug perturbation networks built using QUIZ-C are topologically distinct, reflecting the biological uniqueness of the cell lines in CMap, and are enriched in known drug targets. Pathopticon demonstrates a better prediction performance than solely cheminformatic measures as well as state-of-the-art network and deep learning-based methods. Top predictions made by Pathopticon have high chemical structural diversity, suggesting their potential for building compound libraries. In proof-of-concept applications on vascular diseases, we demonstrate that Pathopticon helps guide in vitro experiments by identifying pathways that are potentially regulated by the predicted therapeutic candidates. CONCLUSIONS Our network-based analytical framework integrating pharmacogenomics and cheminformatics (available at https://github.com/r-duh/Pathopticon ) provides a feasible blueprint for a cell type-specific drug discovery and repositioning platform with broad implications for the efficiency and success of drug development.
Collapse
Affiliation(s)
- Arda Halu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA.
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA.
| | - Sarvesh Chelvanambi
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Julius L Decano
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Joan T Matamalas
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Mary Whelan
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Takaharu Asano
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Namitra Kalicharran
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA
| | - Joseph Loscalzo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Masanori Aikawa
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA.
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Center for Life Sciences Boston Bldg., 17th Floor, 3 Blackfan Street, Boston, MA, 02115, USA.
| |
Collapse
|
3
|
Zhang L, Li J, Zhang Q, Gao J, Zhao K, Asai Y, Hu Z, Gao H. An Integrative analysis of single-cell RNA-seq, transcriptome and Mendelian randomization for the Identification and validation of NAD + Metabolism-Related biomarkers in ulcerative colitis. Int Immunopharmacol 2025; 145:113765. [PMID: 39647286 DOI: 10.1016/j.intimp.2024.113765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/25/2024] [Accepted: 11/30/2024] [Indexed: 12/10/2024]
Abstract
Ulcerative colitis (UC) is a chronic and refractory inflammatory disease of the colon and rectum. This study utilized bioinformatics methods to explore the potential of Nicotinamide adenine dinucleotide (NAD+) metabolism-related genes (NMRGs) as key genes in UC. Using the GSE87466 dataset, differentially expressed NMRGs were identified through differential expression analysis, weighted gene co-expression network analysis (WGCNA), and NMRG scoring. These NMRGs were used as exposure factors, with UC as the outcome, to identify causal candidate genes through Mendelian randomization (MR) analysis. Key genes were further validated as biomarkers using machine learning and expression validation in external datasets (GSE75214, GSE224758). A nomogram based on the expression levels of these biomarkers was constructed to predict UC risk, and the biomarkers' expression was validated through real-time quantitative polymerase chain reaction (RT-qPCR). Subsequently, signaling pathway analysis, enrichment analysis, immune infiltration analysis, and drug prediction were conducted to comprehensively understand the biological roles of the key genes in the human body. Single-cell (GSE116222) and spatial transcriptomic analyses (GSE189184) revealed the expression patterns of these key genes in specific cell types. NCF2, IL1B, S100A8, and SLC26A2 were identified as biomarkers, with NCF2 and IL1B serving as protective factors and S100A8 and SLC26A2 as risk factors for UC. The nomogram based on these biomarkers demonstrated strong predictive value. Functional analysis revealed significant IL1B, NCF2, and S100A8 enrichment in pathways such as IL-4 and IL-13 signaling, while SLC26A2 was strongly associated with respiratory electron transport. Significant differences in immune cells, such as macrophages and neutrophils, were also observed. Single-cell analysis showed high expression of NCF2, IL1B, and S100A8 in monocytes, while SLC26A2 was primarily expressed in epithelial cells, intestinal epithelial cells, and mast cells. Overall, these findings reveal the roles of NMRGs, providing valuable insights into the diagnosis and treatment of UC patients.
Collapse
Affiliation(s)
- Longxiang Zhang
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang, China
| | - Jian Li
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang, China
| | - Qiqi Zhang
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang, China
| | - Jianshu Gao
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang, China
| | - Keke Zhao
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang, China
| | - Yersen Asai
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang, China
| | - Ziying Hu
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang, China
| | - Hongliang Gao
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, Xinjiang, China.
| |
Collapse
|
4
|
Winge MCG, Nasrallah M, Jackrazi LV, Guo KQ, Fuhriman JM, Szafran R, Ramanathan M, Gurevich I, Nguyen NT, Siprashvili Z, Inayathullah M, Rajadas J, Porter DF, Khavari PA, Butte AJ, Marinkovich MP. Repurposing an epithelial sodium channel inhibitor as a therapy for murine and human skin inflammation. Sci Transl Med 2024; 16:eade5915. [PMID: 39661704 DOI: 10.1126/scitranslmed.ade5915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/12/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024]
Abstract
Inflammatory skin disease is characterized by a pathologic interplay between skin cells and immunocytes and can result in disfiguring cutaneous lesions and systemic inflammation. Immunosuppression is commonly used to target the inflammatory component; however, these drugs are often expensive and associated with side effects. To identify previously unidentified targets, we carried out a nonbiased informatics screen to identify drug compounds with an inverse transcriptional signature to keratinocyte inflammatory signals. Using psoriasis, a prototypic inflammatory skin disease, as a model, we used pharmacologic, transcriptomic, and proteomic characterization to find that benzamil, the benzyl derivative of the US Food and Drug Administration-approved diuretic amiloride, effectively reversed keratinocyte-driven inflammatory signaling. Through three independent mouse models of skin inflammation (Rac1G12V transgenic mice, topical imiquimod, and human skin xenografts from patients with psoriasis), we found that benzamil disrupted pathogenic interactions between the small GTPase Rac1 and its adaptor NCK1. This reduced STAT3 and NF-κB signaling and downstream cytokine production in keratinocytes. Genetic knockdown of sodium channels or pharmacological inhibition by benzamil prevented excess Rac1-NCK1 binding and limited proinflammatory signaling pathway activation in patient-derived keratinocytes without systemic immunosuppression. Both systemic and topical applications of benzamil were efficacious, suggesting that it may be a potential therapeutic avenue for treating skin inflammation.
Collapse
Affiliation(s)
- Mårten C G Winge
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mazen Nasrallah
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leandra V Jackrazi
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Konnie Q Guo
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jessica M Fuhriman
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rebecca Szafran
- Unit of Dermatology, ME GHR, Karolinska University Hospital, SE-17176 Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Muthukumar Ramanathan
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Irina Gurevich
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ngon T Nguyen
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zurab Siprashvili
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mohammed Inayathullah
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Jayakumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Douglas F Porter
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paul A Khavari
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Dermatology Service, Veterans Affairs Medical Center, Palo Alto, CA 94304, USA
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - M Peter Marinkovich
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Dermatology Service, Veterans Affairs Medical Center, Palo Alto, CA 94304, USA
| |
Collapse
|
5
|
Yuan H, Hicks P, Ahmadian M, Johnson KA, Valtadoros L, Krishnan A. Annotating publicly-available samples and studies using interpretable modeling of unstructured metadata. Brief Bioinform 2024; 26:bbae652. [PMID: 39710433 DOI: 10.1093/bib/bbae652] [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: 07/16/2024] [Revised: 10/31/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024] Open
Abstract
Reusing massive collections of publicly available biomedical data can significantly impact knowledge discovery. However, these public samples and studies are typically described using unstructured plain text, hindering the findability and further reuse of the data. To combat this problem, we propose txt2onto 2.0, a general-purpose method based on natural language processing and machine learning for annotating biomedical unstructured metadata to controlled vocabularies of diseases and tissues. Compared to the previous version (txt2onto 1.0), which uses numerical embeddings as features, this new version uses words as features, resulting in improved interpretability and performance, especially when few positive training instances are available. Txt2onto 2.0 uses embeddings from a large language model during prediction to deal with unseen-yet-relevant words related to each disease and tissue term being predicted from the input text, thereby explaining the basis of every annotation. We demonstrate the generalizability of txt2onto 2.0 by accurately predicting disease annotations for studies from independent datasets, using proteomics and clinical trials as examples. Overall, our approach can annotate biomedical text regardless of experimental types or sources. Code, data, and trained models are available at https://github.com/krishnanlab/txt2onto2.0.
Collapse
Affiliation(s)
- Hao Yuan
- Genetics and Genome Sciences Program, Michigan State University, East Lansing, MI 48823, United States
- Ecology, Evolution, and Behavior Program, Michigan State University, East Lansing, MI 48823, United States
| | - Parker Hicks
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Mansooreh Ahmadian
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Kayla A Johnson
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Lydia Valtadoros
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Arjun Krishnan
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| |
Collapse
|
6
|
Qi X, Zhao L, Tian C, Li Y, Chen ZL, Huo P, Chen R, Liu X, Wan B, Yang S, Zhao Y. Predicting transcriptional responses to novel chemical perturbations using deep generative model for drug discovery. Nat Commun 2024; 15:9256. [PMID: 39462106 PMCID: PMC11513139 DOI: 10.1038/s41467-024-53457-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Understanding transcriptional responses to chemical perturbations is central to drug discovery, but exhaustive experimental screening of disease-compound combinations is unfeasible. To overcome this limitation, here we introduce PRnet, a perturbation-conditioned deep generative model that predicts transcriptional responses to novel chemical perturbations that have never experimentally perturbed at bulk and single-cell levels. Evaluations indicate that PRnet outperforms alternative methods in predicting responses across novel compounds, pathways, and cell lines. PRnet enables gene-level response interpretation and in-silico drug screening for diseases based on gene signatures. PRnet further identifies and experimentally validates novel compound candidates against small cell lung cancer and colorectal cancer. Lastly, PRnet generates a large-scale integration atlas of perturbation profiles, covering 88 cell lines, 52 tissues, and various compound libraries. PRnet provides a robust and scalable candidate recommendation workflow and successfully recommends drug candidates for 233 diseases. Overall, PRnet is an effective and valuable tool for gene-based therapeutics screening.
Collapse
Affiliation(s)
- Xiaoning Qi
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lianhe Zhao
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chenyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yueyue Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhen-Lin Chen
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Intelligent Information Processing of Chinese Academy of Sciences (CAS), Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| | - Peipei Huo
- Luoyang Institute of Information Technology Industries, Luoyang, Henan, China
| | - Runsheng Chen
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaodong Liu
- University of Chinese Academy Sciences, Nanjing, Jiangsu, China
| | - Baoping Wan
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| | - Shengyong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Yi Zhao
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
7
|
Ganapathiraju MK, Bhatia T, Deshpande S, Wesesky M, Wood J, Nimgaonkar VL. Schizophrenia Interactome-Derived Repurposable Drugs and Randomized Controlled Trials of Two Candidates. Biol Psychiatry 2024; 96:651-658. [PMID: 38950808 DOI: 10.1016/j.biopsych.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/29/2024] [Accepted: 06/09/2024] [Indexed: 07/03/2024]
Abstract
There is a substantial unmet need for effective and patient-acceptable drugs to treat severe mental illnesses such as schizophrenia (SZ). Computational analysis of genomic, transcriptomic, and pharmacologic data generated in the past 2 decades enables repurposing of drugs or compounds with acceptable safety profiles, namely those that are U.S. Food and Drug Administration approved or have reached late stages in clinical trials. We developed a rational approach to achieve this computationally for SZ by studying drugs that target the proteins in its protein interaction network (interactome). This involved contrasting the transcriptomic modulations observed in the disorder and the drug; our analyses resulted in 12 candidate drugs, 9 of which had additional supportive evidence whereby their target networks were enriched for pathways relevant to SZ etiology or for genes that had an association with diseases pathogenically similar to SZ. To translate these computational results to the clinic, these shortlisted drugs must be tested empirically through randomized controlled trials, in which their previous safety approvals obviate the need for time-consuming phase 1 and 2 studies. We selected 2 among the shortlisted candidates based on likely adherence and side-effect profiles. We are testing them through adjunctive randomized controlled trials for patients with SZ or schizoaffective disorder who experienced incomplete resolution of psychotic features with conventional treatment. The integrated computational analysis for identifying and ranking drugs for clinical trials can be iterated as additional data are obtained. Our approach could be expanded to enable disease subtype-specific drug discovery in the future and should also be exploited for other psychiatric disorders.
Collapse
Affiliation(s)
- Madhavi K Ganapathiraju
- Department of Biomedical Informatics and Intelligent Systems Program, University of Pittsburgh, Pittsburgh, Pennsylvania; Carnegie Mellon University in Qatar, Doha, Qatar.
| | - Triptish Bhatia
- Department of Psychiatry, Centre of Excellence in Mental Health, ABVIMS - Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Smita Deshpande
- Department of Psychiatry, St John's Medical College Hospital, Koramangala, Bengaluru, Karnataka, India
| | - Maribeth Wesesky
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joel Wood
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Vishwajit L Nimgaonkar
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Veterans Administration Pittsburgh Healthcare System, Pittsburgh, Pennsylvania.
| |
Collapse
|
8
|
Zhou X, Xin G, Wan C, Li F, Wang Y, Zhang K, Yu X, Li S, Huang W. Myricetin reduces platelet PANoptosis in sepsis to delay disseminated intravascular coagulation. Biochem Biophys Res Commun 2024; 724:150140. [PMID: 38852506 DOI: 10.1016/j.bbrc.2024.150140] [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: 05/01/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/11/2024]
Abstract
Sepsis is a severe inflammatory disease characterized by cytokine storm, often accompanied by disseminated intravascular coagulation (DIC). PANoptosis is a novel form of cell death triggered by cytokine storms, characterized by a cascade reaction of pyroptosis, apoptosis, and necroptosis. It exists in septic platelets and is closely associated with the onset and progression of DIC. However, there remains an unmet need for drugs targeting PANoptosis. The anti-PANoptosis effect of myricetin was predicted using network pharmacology and confirmed through molecular docking. In vitro platelet activation models demonstrated that myricetin significantly attenuated platelet particle release, integrin activation, adhesion, spreading, clot retraction, and aggregation. Moreover, in a sepsis model, myricetin reduced inflammatory infiltration in lung tissue and platelet activation while improving DIC. Additionally, whole blood sequencing samples from sepsis patients and healthy individuals were analyzed to elucidate the up-regulation of the PANoptosis targets. Our findings demonstrate the inhibitory effect of myricetin on septic platelet PANoptosis, indicating its potential as a novel anti-cellular PANoptosis candidate and therapeutic agent for septic DIC. Furthermore, our study establishes a foundation for utilizing network pharmacology in the discovery of new drugs to treat various diseases.
Collapse
Affiliation(s)
- Xiaoli Zhou
- Natural and Biomimetic Medicine Research Center, West China School of Medicine, West China Hospital, Sichuan University, China; College of Health, Yuncheng Vocational and Technical University, China
| | - Guang Xin
- Natural and Biomimetic Medicine Research Center, West China School of Medicine, West China Hospital, Sichuan University, China
| | - Chengyu Wan
- Natural and Biomimetic Medicine Research Center, West China School of Medicine, West China Hospital, Sichuan University, China
| | - Fan Li
- Natural and Biomimetic Medicine Research Center, West China School of Medicine, West China Hospital, Sichuan University, China
| | - Yilan Wang
- Natural and Biomimetic Medicine Research Center, West China School of Medicine, West China Hospital, Sichuan University, China
| | - Kun Zhang
- Natural and Biomimetic Medicine Research Center, West China School of Medicine, West China Hospital, Sichuan University, China
| | - Xiuxian Yu
- Natural and Biomimetic Medicine Research Center, West China School of Medicine, West China Hospital, Sichuan University, China
| | - Shiyi Li
- Natural and Biomimetic Medicine Research Center, West China School of Medicine, West China Hospital, Sichuan University, China
| | - Wen Huang
- Natural and Biomimetic Medicine Research Center, West China School of Medicine, West China Hospital, Sichuan University, China.
| |
Collapse
|
9
|
Kataria A, Srivastava A, Singh DD, Haque S, Han I, Yadav DK. Systematic computational strategies for identifying protein targets and lead discovery. RSC Med Chem 2024; 15:2254-2269. [PMID: 39026640 PMCID: PMC11253860 DOI: 10.1039/d4md00223g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/10/2024] [Indexed: 07/20/2024] Open
Abstract
Computational algorithms and tools have retrenched the drug discovery and development timeline. The applicability of computational approaches has gained immense relevance owing to the dramatic surge in the structural information of biomacromolecules and their heteromolecular complexes. Computational methods are now extensively used in identifying new protein targets, druggability assessment, pharmacophore mapping, molecular docking, the virtual screening of lead molecules, bioactivity prediction, molecular dynamics of protein-ligand complexes, affinity prediction, and for designing better ligands. Herein, we provide an overview of salient components of recently reported computational drug-discovery workflows that includes algorithms, tools, and databases for protein target identification and optimized ligand selection.
Collapse
Affiliation(s)
- Arti Kataria
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) Hamilton MT 59840 USA
| | - Ankit Srivastava
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) Hamilton MT 59840 USA
| | - Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan Jaipur India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University Jazan-45142 Saudi Arabia
| | - Ihn Han
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical & Biological Physics, Kwangwoon University Seoul 01897 Republic of Korea +82 32 820 4948
| | - Dharmendra Kumar Yadav
- Department of Biologics, College of Pharmacy, Gachon University Hambakmoeiro 191, Yeonsu-gu Incheon 21924 Republic of Korea
| |
Collapse
|
10
|
Cai K, Zhu Y, Zheng Y, Wang H, Qian Y. B2R-Targeting Radiotracer for PET/MR Imaging of Hepatocellular Carcinoma and Guiding Anti-B2R Therapy. ACS Med Chem Lett 2024; 15:1080-1087. [PMID: 39015273 PMCID: PMC11247633 DOI: 10.1021/acsmedchemlett.4c00155] [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: 04/08/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
The bradykinin B2 receptor (B2R) is overexpressed in a wide variety of tumors and is a well-defined target for tumor imaging and therapy. The hybrid positron emission tomography/magnetic resonance imaging (PET/MRI) scanner is considered a noninvasive and advanced instrument for precise tumor imaging. In this work, we developed a novel B2R-targeting radiotracer, 68Ga-DOTA-icatibant, for quantifying B2R expression. 68Ga-DOTA-icatibant showed high stability, fast clearance and specific binding to B2R. PET/MR imaging revealed excellent tumor accumulation, and the uptake in tumors could be blocked by DOTA-icatibant. Icatibant-mediated anti-B2R therapy downregulated B2R expression in tumor cells and inhibited the growth of HepG2 tumors, and the decrease in tumor uptake was monitored by timely PET/MR imaging. Hematoxylin and eosin (H&E) and immunohistochemical staining results further demonstrated that the efficacy of anti-B2R could be accurately monitored with the developed PET/MR imaging radiotracer. 68Ga-DOTA-icatibant can be utilized to noninvasively determine B2R expression and dynamically and sensitively monitor the efficacy of anti-B2R therapy.
Collapse
Affiliation(s)
- Ke Cai
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
| | - Yunzhu Zhu
- Department
of Infectious Diseases, The First Affiliated
Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
| | - Yifan Zheng
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
| | - Hui Wang
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
| | - Yinfeng Qian
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
- Department
of Radiology, The First Affiliated Hospital
of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
| |
Collapse
|
11
|
Honap S, Jairath V, Danese S, Peyrin-Biroulet L. Navigating the complexities of drug development for inflammatory bowel disease. Nat Rev Drug Discov 2024; 23:546-562. [PMID: 38778181 DOI: 10.1038/s41573-024-00953-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2024] [Indexed: 05/25/2024]
Abstract
Inflammatory bowel disease (IBD) - consisting of ulcerative colitis and Crohn's disease - is a complex, heterogeneous, immune-mediated inflammatory condition with a multifactorial aetiopathogenesis. Despite therapeutic advances in this arena, a ceiling effect has been reached with both single-agent monoclonal antibodies and advanced small molecules. Therefore, there is a need to identify novel targets, and the development of companion biomarkers to select responders is vital. In this Perspective, we examine how advances in machine learning and tissue engineering could be used at the preclinical stage where attrition rates are high. For novel agents reaching clinical trials, we explore factors decelerating progression, particularly the decline in IBD trial recruitment, and assess how innovative approaches such as reconfiguring trial designs, harmonizing end points and incorporating digital technologies into clinical trials can address this. Harnessing opportunities at each stage of the drug development process may allow for incremental gains towards more effective therapies.
Collapse
Affiliation(s)
- Sailish Honap
- Department of Gastroenterology, St George's University Hospitals NHS Foundation Trust, London, UK.
- School of Immunology and Microbial Sciences, King's College London, London, UK.
- INFINY Institute, Nancy University Hospital, Vandœuvre-lès-Nancy, France.
| | - Vipul Jairath
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University, London, Ontario, Canada
- Lawson Health Research Institute, Western University, London, Ontario, Canada
- Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada
| | - Silvio Danese
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Laurent Peyrin-Biroulet
- INFINY Institute, Nancy University Hospital, Vandœuvre-lès-Nancy, France.
- Department of Gastroenterology, Nancy University Hospital, Vandœuvre-lès-Nancy, France.
- INSERM, NGERE, University of Lorraine, Nancy, France.
- FHU-CURE, Nancy University Hospital, Vandœuvre-lès-Nancy, France.
- Groupe Hospitalier privé Ambroise Paré - Hartmann, Paris IBD Center, Neuilly sur Seine, France.
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
12
|
Karunakaran KB, Ganapathiraju MK. Malignant peritoneal mesothelioma interactome with 417 novel protein-protein interactions. BJC REPORTS 2024; 2:42. [PMID: 39516360 PMCID: PMC11524009 DOI: 10.1038/s44276-024-00062-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Malignant peritoneal mesothelioma (MPeM) is an aggressive cancer affecting the abdominal peritoneal lining and intra-abdominal organs, with a median survival of ~2.5 years. METHODS We constructed the protein interactome of 59 MPeM-associated genes with previously known protein-protein interactions (PPIs) as well as novel PPIs predicted using our previously developed HiPPIP computational model and analysed it for transcriptomic and functional associations and for repurposable drugs. RESULTS The MPeM interactome had over 400 computationally predicted PPIs and 4700 known PPIs. Transcriptomic evidence validated 75.6% of the genes in the interactome and 65% of the novel interactors. Some genes had tissue-specific expression in extramedullary hematopoietic sites and the expression of some genes could be correlated with unfavourable prognoses in various cancers. 39 out of 152 drugs that target the proteins in the interactome were identified as potentially repurposable for MPeM, with 29 having evidence from prior clinical trials, animal models or cell lines for effectiveness against peritoneal and pleural mesothelioma and primary peritoneal cancer. Functional modules related to chromosomal segregation, transcriptional dysregulation, IL-6 production and hematopoiesis were identified from the interactome. The MPeM interactome overlapped significantly with the malignant pleural mesothelioma interactome, revealing shared molecular pathways. CONCLUSIONS Our findings demonstrate the utility of the interactome in uncovering biological associations and in generating clinically translatable results.
Collapse
Affiliation(s)
- Kalyani B Karunakaran
- Supercomputer Education and Research Centre, Indian Institute of Science, Bengaluru, 560012, India.
| | - Madhavi K Ganapathiraju
- Department of Biomedical Informatics, School of Medicine, and Intelligent Systems Program, School of Computing and Information, University of Pittsburgh, 5607 Baum Blvd, 5th Floor, Pittsburgh, PA, 15206, USA.
- Carnegie Mellon University in Qatar, Doha, Qatar.
| |
Collapse
|
13
|
Bui NL, Hoang DA, Ho QA, Nguyen Thi TN, Singh V, Chu DT. Drug repurposing for metabolic disorders: Scientific, technological and economic issues. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:321-336. [PMID: 38942542 DOI: 10.1016/bs.pmbts.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Obesity, diabetes, and other metabolic disorders place a huge burden on both the physical health and financial well-being of the community. While the need for effective treatment of metabolic disorders remains urgent and the reality is that traditional drug development involves high costs and a very long time with many pre-clinical and clinical trials, the need for drug repurposing has emerged as a potential alternative. Scientific evidence has shown the anti-diabetic and anti-obesity effects of old drugs, which were initially utilized for the treatment of inflammation, depression, infections, and even cancers. The drug library used modern technological methods to conduct drug screening. Computational molecular docking, genome-wide association studies, or omics data mining are advantageous and unavoidable methods for drug repurposing. Drug repurposing offers a promising avenue for economic efficiency in healthcare, especially for less common metabolic diseases, despite the need for rigorous research and validation. In this chapter, we aim to explore the scientific, technological, and economic issues surrounding drug repurposing for metabolic disorders. We hope to shed light on the potential of this approach and the challenges that need to be addressed to make it a viable option in the treatment of metabolic disorders, especially in the future fight against metabolic disorders.
Collapse
Affiliation(s)
- Nhat-Le Bui
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Duc-Anh Hoang
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
| | - Quang-Anh Ho
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
| | - Thao-Nguyen Nguyen Thi
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, India
| | - Dinh-Toi Chu
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam.
| |
Collapse
|
14
|
Loganathan T, Fletcher J, Abraham P, Kannangai R, Chakraborty C, El Allali A, Alsamman AM, Zayed H, C GPD. Expression analysis and mapping of Viral-Host Protein interactions of Poxviridae suggests a lead candidate molecule targeting Mpox. BMC Infect Dis 2024; 24:483. [PMID: 38730352 PMCID: PMC11088078 DOI: 10.1186/s12879-024-09332-x] [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: 09/18/2023] [Accepted: 04/18/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Monkeypox (Mpox) is an important human pathogen without etiological treatment. A viral-host interactome study may advance our understanding of molecular pathogenesis and lead to the discovery of suitable therapeutic targets. METHODS GEO Expression datasets characterizing mRNA profile changes in different host responses to poxviruses were analyzed for shared pathway identification, and then, the Protein-protein interaction (PPI) maps were built. The viral gene expression datasets of Monkeypox virus (MPXV) and Vaccinia virus (VACV) were used to identify the significant viral genes and further investigated for their binding to the library of targeting molecules. RESULTS Infection with MPXV interferes with various cellular pathways, including interleukin and MAPK signaling. While most host differentially expressed genes (DEGs) are predominantly downregulated upon infection, marked enrichments in histone modifiers and immune-related genes were observed. PPI analysis revealed a set of novel virus-specific protein interactions for the genes in the above functional clusters. The viral DEGs exhibited variable expression patterns in three studied cell types: primary human monocytes, primary human fibroblast, and HeLa, resulting in 118 commonly deregulated proteins. Poxvirus proteins C6R derived protein K7 and K7R of MPXV and VACV were prioritized as targets for potential therapeutic interventions based on their histone-regulating and immunosuppressive properties. In the computational docking and Molecular Dynamics (MD) experiments, these proteins were shown to bind the candidate small molecule S3I-201, which was further prioritized for lead development. RESULTS MPXV circumvents cellular antiviral defenses by engaging histone modification and immune evasion strategies. C6R-derived protein K7 binding candidate molecule S3I-201 is a priority promising candidate for treating Mpox.
Collapse
Affiliation(s)
- Tamizhini Loganathan
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore-632014, Tamil Nadu, India
| | - John Fletcher
- Department of Clinical Virology, Christian Medical College, Tamil Nadu, Vellore, 632004, India
| | - Priya Abraham
- Department of Clinical Virology, Christian Medical College, Tamil Nadu, Vellore, 632004, India
| | - Rajesh Kannangai
- Department of Clinical Virology, Christian Medical College, Tamil Nadu, Vellore, 632004, India
| | | | - Achraf El Allali
- Bioinformatics Laboratory, College of Computing, Mohammed VI Polytechnic University, Ben Guerir, Mohammed, Morocco.
| | - Alsamman M Alsamman
- Department of Genome Mapping, Molecular Genetics, and Genome Mapping Laboratory, Agricultural Genetic Engineering Research Institute, Giza, Egypt
| | - Hatem Zayed
- Department of Biomedical Sciences College of Health Sciences, QU. Health, Qatar University, Doha, Qatar
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore-632014, Tamil Nadu, India.
| |
Collapse
|
15
|
Haderk F, Chou YT, Cech L, Fernández-Méndez C, Yu J, Olivas V, Meraz IM, Barbosa Rabago D, Kerr DL, Gomez C, Allegakoen DV, Guan J, Shah KN, Herrington KA, Gbenedio OM, Nanjo S, Majidi M, Tamaki W, Pourmoghadam YK, Rotow JK, McCoach CE, Riess JW, Gutkind JS, Tang TT, Post L, Huang B, Santisteban P, Goodarzi H, Bandyopadhyay S, Kuo CJ, Roose JP, Wu W, Blakely CM, Roth JA, Bivona TG. Focal adhesion kinase-YAP signaling axis drives drug-tolerant persister cells and residual disease in lung cancer. Nat Commun 2024; 15:3741. [PMID: 38702301 PMCID: PMC11068778 DOI: 10.1038/s41467-024-47423-0] [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: 01/04/2022] [Accepted: 03/18/2024] [Indexed: 05/06/2024] Open
Abstract
Targeted therapy is effective in many tumor types including lung cancer, the leading cause of cancer mortality. Paradigm defining examples are targeted therapies directed against non-small cell lung cancer (NSCLC) subtypes with oncogenic alterations in EGFR, ALK and KRAS. The success of targeted therapy is limited by drug-tolerant persister cells (DTPs) which withstand and adapt to treatment and comprise the residual disease state that is typical during treatment with clinical targeted therapies. Here, we integrate studies in patient-derived and immunocompetent lung cancer models and clinical specimens obtained from patients on targeted therapy to uncover a focal adhesion kinase (FAK)-YAP signaling axis that promotes residual disease during oncogenic EGFR-, ALK-, and KRAS-targeted therapies. FAK-YAP signaling inhibition combined with the primary targeted therapy suppressed residual drug-tolerant cells and enhanced tumor responses. This study unveils a FAK-YAP signaling module that promotes residual disease in lung cancer and mechanism-based therapeutic strategies to improve tumor response.
Collapse
Affiliation(s)
- Franziska Haderk
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Yu-Ting Chou
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Lauren Cech
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Celia Fernández-Méndez
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científícas (CSIC) y Universidad Autónoma de Madrid (UAM), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Johnny Yu
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
| | - Victor Olivas
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Ismail M Meraz
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dora Barbosa Rabago
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - D Lucas Kerr
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Carlos Gomez
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - David V Allegakoen
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Juan Guan
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Khyati N Shah
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kari A Herrington
- Center for Advanced Light Microscopy, University of California, San Francisco, San Francisco, CA, USA
| | | | - Shigeki Nanjo
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Mourad Majidi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Whitney Tamaki
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Yashar K Pourmoghadam
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Julia K Rotow
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Caroline E McCoach
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan W Riess
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - J Silvio Gutkind
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Tracy T Tang
- Vivace Therapeutics, Inc., 1500 Fashion Island Blvd., Suite 102, San Mateo, CA, USA
| | - Leonard Post
- Vivace Therapeutics, Inc., 1500 Fashion Island Blvd., Suite 102, San Mateo, CA, USA
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científícas (CSIC) y Universidad Autónoma de Madrid (UAM), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Hani Goodarzi
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
| | - Sourav Bandyopadhyay
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeroen P Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Wei Wu
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Collin M Blakely
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
16
|
Oskotsky TT, Bhoja A, Bunis D, Le BL, Tang AS, Kosti I, Li C, Houshdaran S, Sen S, Vallvé-Juanico J, Wang W, Arthurs E, Govil A, Mahoney L, Lang L, Gaudilliere B, Stevenson DK, Irwin JC, Giudice LC, McAllister SL, Sirota M. Identifying therapeutic candidates for endometriosis through a transcriptomics-based drug repositioning approach. iScience 2024; 27:109388. [PMID: 38510116 PMCID: PMC10952035 DOI: 10.1016/j.isci.2024.109388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/29/2023] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Existing medical treatments for endometriosis-related pain are often ineffective, underscoring the need for new therapeutic strategies. In this study, we applied a computational drug repurposing pipeline to stratified and unstratified disease signatures based on endometrial gene expression data to identify potential therapeutics from existing drugs, based on expression reversal. Of 3,131 unique genes differentially expressed by at least one of six endometriosis signatures, only 308 (9.8%) were in common; however, 221 out of 299 drugs identified, (73.9%) were shared. We selected fenoprofen, an uncommonly prescribed NSAID that was the top therapeutic candidate for further investigation. When testing fenoprofen in an established rat model of endometriosis, fenoprofen successfully alleviated endometriosis-associated vaginal hyperalgesia, a surrogate marker for endometriosis-related pain. These findings validate fenoprofen as a therapeutic that could be utilized more frequently for endometriosis and suggest the utility of the aforementioned computational drug repurposing approach for endometriosis.
Collapse
Affiliation(s)
- Tomiko T. Oskotsky
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| | - Arohee Bhoja
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
- Carnegie Mellon University, Pittsburgh, PA, USA
| | - Daniel Bunis
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| | - Brian L. Le
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| | - Alice S. Tang
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| | - Idit Kosti
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| | - Christine Li
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
| | - Sahar Houshdaran
- Department of Obstetrics, Gynecology and Reproductive Sciences, UCSF, San Francisco, CA, USA
| | - Sushmita Sen
- Department of Obstetrics, Gynecology and Reproductive Sciences, UCSF, San Francisco, CA, USA
| | - Júlia Vallvé-Juanico
- Department of Obstetrics, Gynecology and Reproductive Sciences, UCSF, San Francisco, CA, USA
| | - Wanxin Wang
- Department of Obstetrics, Gynecology and Reproductive Sciences, UCSF, San Francisco, CA, USA
| | - Erin Arthurs
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Arpita Govil
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Lauren Mahoney
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Lindsey Lang
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Brice Gaudilliere
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University, Stanford, CA, USA
| | | | - Juan C. Irwin
- Department of Obstetrics, Gynecology and Reproductive Sciences, UCSF, San Francisco, CA, USA
| | - Linda C. Giudice
- Department of Obstetrics, Gynecology and Reproductive Sciences, UCSF, San Francisco, CA, USA
| | | | - Marina Sirota
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| |
Collapse
|
17
|
Israr J, Alam S, Singh V, Kumar A. Repurposing of biologics and biopharmaceuticals. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 205:277-302. [PMID: 38789184 DOI: 10.1016/bs.pmbts.2024.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
The field of drug repurposing is gaining attention as a way to introduce pharmaceutical agents with established safety profiles to new patient populations. This approach involves finding new applications for existing drugs through observations or deliberate efforts to understand their mechanisms of action. Recent advancements in bioinformatics and pharmacology, along with the availability of extensive data repositories and analytical techniques, have fueled the demand for novel methodologies in pharmaceutical research and development. To facilitate systematic drug repurposing, various computational methodologies have emerged, combining experimental techniques and in silico approaches. These methods have revolutionized the field of drug discovery by enabling the efficient repurposing of screens. However, establishing an ideal drug repurposing pipeline requires the integration of molecular data accessibility, analytical proficiency, experimental design expertise, and a comprehensive understanding of clinical development processes. This chapter explores the key methodologies used in systematic drug repurposing and discusses the stakeholders involved in this field. It emphasizes the importance of strategic alliances to enhance the success of repurposing existing compounds for new indications. Additionally, the chapter highlights the current benefits, considerations, and challenges faced in the repurposing process, which is pursued by both biotechnology and pharmaceutical companies. Overall, drug repurposing holds great promise in expanding the use of existing drugs and bringing them to new patient populations. With the advancements in computational methodologies and the collaboration of various stakeholders, this approach has the potential to accelerate drug development and improve patient outcomes.
Collapse
Affiliation(s)
- Juveriya Israr
- Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Lucknow-Deva Road, Barabanki, Uttar Pradesh, India; Department of Biotechnology Era University, Lucknow, Uttar Pradesh, India
| | - Shabroz Alam
- Department of Biotechnology Era University, Lucknow, Uttar Pradesh, India
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| | - Ajay Kumar
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University, Mandhana, Kanpur, Uttar Pradesh, India.
| |
Collapse
|
18
|
Habib M, Lalagkas PN, Melamed RD. Mapping drug biology to disease genetics to discover drug impacts on the human phenome. BIOINFORMATICS ADVANCES 2024; 4:vbae038. [PMID: 38736684 PMCID: PMC11087821 DOI: 10.1093/bioadv/vbae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/18/2024] [Accepted: 03/07/2024] [Indexed: 05/14/2024]
Abstract
Motivation Medications can have unexpected effects on disease, including not only harmful drug side effects, but also beneficial drug repurposing. These effects on disease may result from hidden influences of drugs on disease gene networks. Then, discovering how biological effects of drugs relate to disease biology can both provide insight into the mechanism of latent drug effects, and can help predict new effects. Results Here, we develop Draphnet, a model that integrates molecular data on 429 drugs and gene associations of nearly 200 common phenotypes to learn a network that explains drug effects on disease in terms of these molecular signals. We present evidence that our method can both predict drug effects, and can provide insight into the biology of unexpected drug effects on disease. Using Draphnet to map a drug's known molecular effects to downstream effects on the disease genome, we put forward disease genes impacted by drugs, and we suggest a new grouping of drugs based on shared effects on the disease genome. Our approach has multiple applications, including predicting drug uses and learning drug biology, with implications for personalized medicine. Availability and implementation Code to reproduce the analysis is available at https://github.com/RDMelamed/drug-phenome.
Collapse
Affiliation(s)
- Mamoon Habib
- Department of Computer Science, University of Massachusetts Lowell, Lowell, MA 01854, United States
| | | | - Rachel D Melamed
- Department of Biological Science, University of Massachusetts Lowell, Lowell, MA 01854, United States
| |
Collapse
|
19
|
Xie J, Rao J, Xie J, Zhao H, Yang Y. Predicting disease-gene associations through self-supervised mutual infomax graph convolution network. Comput Biol Med 2024; 170:108048. [PMID: 38310804 DOI: 10.1016/j.compbiomed.2024.108048] [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: 09/04/2023] [Revised: 12/19/2023] [Accepted: 01/26/2024] [Indexed: 02/06/2024]
Abstract
Illuminating associations between diseases and genes can help reveal the pathogenesis of syndromes and contribute to treatments, but a large number of associations remained unexplored. To identify novel disease-gene associations, many computational methods have been developed using disease and gene-related prior knowledge. However, these methods remain of relatively inferior performance due to the limited external data sources and the inevitable noise among the prior knowledge. In this study, we have developed a new method, Self-Supervised Mutual Infomax Graph Convolution Network (MiGCN), to predict disease-gene associations under the guidance of external disease-disease and gene-gene collaborative graphs. The noises within the collaborative graphs were eliminated by maximizing the mutual information between nodes and neighbors through a graphical mutual infomax layer. In parallel, the node interactions were strengthened by a novel informative message passing layer to improve the learning ability of graph neural network. The extensive experiments showed that our model achieved performance improvement over the state-of-art method by more than 8 % on AUC. The datasets, source codes and trained models of MiGCN are available at https://github.com/biomed-AI/MiGCN.
Collapse
Affiliation(s)
- Jiancong Xie
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510000, China
| | - Jiahua Rao
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510000, China
| | - Junjie Xie
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510000, China
| | - Huiying Zhao
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510000, China.
| | - Yuedong Yang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510000, China.
| |
Collapse
|
20
|
Weth FR, Hoggarth GB, Weth AF, Paterson E, White MPJ, Tan ST, Peng L, Gray C. Unlocking hidden potential: advancements, approaches, and obstacles in repurposing drugs for cancer therapy. Br J Cancer 2024; 130:703-715. [PMID: 38012383 PMCID: PMC10912636 DOI: 10.1038/s41416-023-02502-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/30/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023] Open
Abstract
High rates of failure, exorbitant costs, and the sluggish pace of new drug discovery and development have led to a growing interest in repurposing "old" drugs to treat both common and rare diseases, particularly cancer. Cancer, a complex and heterogeneous disease, often necessitates a combination of different treatment modalities to achieve optimal outcomes. The intrinsic polygenicity of cancer, intricate biological signalling networks, and feedback loops make the inhibition of a single target frequently insufficient for achieving the desired therapeutic impact. As a result, addressing these complex or "smart" malignancies demands equally sophisticated treatment strategies. Combinatory treatments that target the multifaceted oncogenic signalling network hold immense promise. Repurposed drugs offer a potential solution to this challenge, harnessing known compounds for new indications. By avoiding the prohibitive costs and long development timelines associated with novel cancer drugs, this approach holds the potential to usher in more effective, efficient, and cost-effective cancer treatments. The pursuit of combinatory therapies through drug repurposing may hold the key to achieving superior outcomes for cancer patients. However, drug repurposing faces significant commercial, technological and regulatory challenges that need to be addressed. This review explores the diverse approaches employed in drug repurposing, delves into the challenges faced by the drug repurposing community, and presents innovative solutions to overcome these obstacles. By emphasising the significance of combinatory treatments within the context of drug repurposing, we aim to unlock the full potential of this approach for enhancing cancer therapy. The positive aspects of drug repurposing in oncology are underscored here; encompassing personalized treatment, accelerated development, market opportunities for shelved drugs, cancer prevention, expanded patient reach, improved patient access, multi-partner collaborations, increased likelihood of approval, reduced costs, and enhanced combination therapy.
Collapse
Affiliation(s)
- Freya R Weth
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand
| | - Georgia B Hoggarth
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
| | - Anya F Weth
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
| | - Erin Paterson
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
| | | | - Swee T Tan
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Lower Hutt, 5040, New Zealand
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Lifeng Peng
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand
| | - Clint Gray
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand.
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| |
Collapse
|
21
|
Mecham A, Stephenson A, Quinteros BI, Brown GS, Piccolo SR. TidyGEO: preparing analysis-ready datasets from Gene Expression Omnibus. J Integr Bioinform 2024; 21:jib-2023-0021. [PMID: 38047898 PMCID: PMC11294518 DOI: 10.1515/jib-2023-0021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 11/20/2023] [Indexed: 12/05/2023] Open
Abstract
TidyGEO is a Web-based tool for downloading, tidying, and reformatting data series from Gene Expression Omnibus (GEO). As a freely accessible repository with data from over 6 million biological samples across more than 4000 organisms, GEO provides diverse opportunities for secondary research. Although scientists may find assay data relevant to a given research question, most analyses require sample-level annotations. In GEO, such annotations are stored alongside assay data in delimited, text-based files. However, the structure and semantics of the annotations vary widely from one series to another, and many annotations are not useful for analysis purposes. Thus, every GEO series must be tidied before it is analyzed. Manual approaches may be used, but these are error prone and take time away from other research tasks. Custom computer scripts can be written, but many scientists lack the computational expertise to create such scripts. To address these challenges, we created TidyGEO, which supports essential data-cleaning tasks for sample-level annotations, such as selecting informative columns, renaming columns, splitting or merging columns, standardizing data values, and filtering samples. Additionally, users can integrate annotations with assay data, restructure assay data, and generate code that enables others to reproduce these steps.
Collapse
Affiliation(s)
- Avery Mecham
- Department of Biology, Brigham Young University, Provo, UT, 84602, USA
| | - Ashlie Stephenson
- Department of Biology, Brigham Young University, Provo, UT, 84602, USA
| | - Badi I. Quinteros
- Department of Biology, Brigham Young University, Provo, UT, 84602, USA
| | - Grace S. Brown
- Department of Biology, Brigham Young University, Provo, UT, 84602, USA
| | | |
Collapse
|
22
|
Gao Z, Ding P, Xu R. IUPHAR review - Data-driven computational drug repurposing approaches for opioid use disorder. Pharmacol Res 2024; 199:106960. [PMID: 37832859 DOI: 10.1016/j.phrs.2023.106960] [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: 09/19/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
Opioid Use Disorder (OUD) is a chronic and relapsing condition characterized by the misuse of opioid drugs, causing significant morbidity and mortality in the United States. Existing medications for OUD are limited, and there is an immediate need to discover treatments with enhanced safety and efficacy. Drug repurposing aims to find new indications for existing medications, offering a time-saving and cost-efficient alternative strategy to traditional drug discovery. Computational approaches have been developed to further facilitate the drug repurposing process. In this paper, we reviewed state-of-the-art data-driven computational drug repurposing approaches for OUD and discussed their advantages and potential challenges. We also highlighted promising repurposed candidate drugs for OUD that were identified by computational drug repurposing techniques and reviewed studies supporting their potential mechanisms of action in treating OUD.
Collapse
Affiliation(s)
- Zhenxiang Gao
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Pingjian Ding
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
23
|
Roberts JA, Varma VR, Jones A, Thambisetty M. Drug Repurposing for Effective Alzheimer's Disease Medicines: Existing Methods and Novel Pharmacoepidemiological Approaches. J Alzheimers Dis 2024; 101:S299-S315. [PMID: 39422962 DOI: 10.3233/jad-240680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Drug repurposing is a methodology used to identify new clinical indications for existing drugs developed for other indications and has been successfully applied in the treatment of numerous conditions. Alzheimer's disease (AD) may be particularly well-suited to the application of drug repurposing methods given the absence of effective therapies and abundance of multi-omic data that has been generated in AD patients recently that may facilitate discovery of candidate AD drugs. A recent focus of drug repurposing has been in the application of pharmacoepidemiologic approaches to drug evaluation. Here, real-world clinical datasets with large numbers of patients are leveraged to establish observational efficacy of candidate drugs for further evaluation in disease models and clinical trials. In this review, we provide a selected overview of methods for drug repurposing, including signature matching, network analysis, molecular docking, phenotypic screening, semantic network, and pharmacoepidemiological analyses. Numerous methods have also been applied specifically to AD with the aim of nominating novel drug candidates for evaluation. These approaches, however, are prone to numerous limitations and potential biases that we have sought to address in the Drug Repurposing for Effective Alzheimer's Medicines (DREAM) study, a multi-step framework for selection and validation of potential drug candidates that has demonstrated the promise of STAT3 inhibitors and re-evaluated evidence for other drug candidates, such as phosphodiesterase inhibitors. Taken together, drug repurposing holds significant promise for development of novel AD therapeutics, particularly as the pace of data generation and development of analytical methods continue to accelerate.
Collapse
Affiliation(s)
- Jackson A Roberts
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, Massachusetts General Brigham, Boston, MA, USA
| | - Vijay R Varma
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Attila Jones
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
24
|
Mokhtari M, Khoshbakht S, Akbari ME, Moravveji SS. BMC3PM: bioinformatics multidrug combination protocol for personalized precision medicine and its application in cancer treatment. BMC Med Genomics 2023; 16:328. [PMID: 38087279 PMCID: PMC10717810 DOI: 10.1186/s12920-023-01745-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND In recent years, drug screening has been one of the most significant challenges in the field of personalized medicine, particularly in cancer treatment. However, several new platforms have been introduced to address this issue, providing reliable solutions for personalized drug validation and safety testing. In this study, we developed a personalized drug combination protocol as the primary input to such platforms. METHODS To achieve this, we utilized data from whole-genome expression profiles of 6173 breast cancer patients, 312 healthy individuals, and 691 drugs. Our approach involved developing an individual pattern of perturbed gene expression (IPPGE) for each patient, which was used as the basis for drug selection. An algorithm was designed to extract personalized drug combinations by comparing the IPPGE and drug signatures. Additionally, we employed the concept of drug repurposing, searching for new benefits of existing drugs that may regulate the desired genes. RESULTS Our study revealed that drug combinations obtained from both specialized and non-specialized cancer medicines were more effective than those extracted from only specialized medicines. Furthermore, we observed that the individual pattern of perturbed gene expression (IPPGE) was unique to each patient, akin to a fingerprint. CONCLUSIONS The personalized drug combination protocol developed in this study offers a methodological interface between drug repurposing and combination drug therapy in cancer treatment. This protocol enables personalized drug combinations to be extracted from hundreds of drugs and thousands of drug combinations, potentially offering more effective treatment options for cancer patients.
Collapse
Affiliation(s)
- Majid Mokhtari
- Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran.
| | - Samane Khoshbakht
- Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran
- Duke Molecular Physiology Institute, Duke University School of Medicine-Cardiology, Durham, NC, 27701, USA
| | | | - Sayyed Sajjad Moravveji
- Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran
| |
Collapse
|
25
|
Lacruz-Pleguezuelos B, Piette O, Garranzo M, Pérez-Serrano D, Milešević J, Espinosa-Salinas I, Ramírez de Molina A, Laguna T, Carrillo de Santa Pau E. FooDrugs: a comprehensive food-drug interactions database with text documents and transcriptional data. Database (Oxford) 2023; 2023:baad075. [PMID: 37951712 PMCID: PMC10640380 DOI: 10.1093/database/baad075] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/15/2023] [Accepted: 10/11/2023] [Indexed: 11/14/2023]
Abstract
Food-drug interactions (FDIs) occur when a food item alters the pharmacokinetics or pharmacodynamics of a drug. FDIs can be clinically relevant, as they can hamper or enhance the therapeutic effects of a drug and impact both their efficacy and their safety. However, knowledge of FDIs in clinical practice is limited. This is partially due to the lack of resources focused on FDIs. Here, we describe FooDrugs, a database that centralizes FDI knowledge retrieved from two different approaches: a natural processing language pipeline that extracts potential FDIs from scientific documents and clinical trials and a molecular similarity approach based on the comparison of gene expression alterations caused by foods and drugs. FooDrugs database stores a total of 3 430 062 potential FDIs, with 1 108 429 retrieved from scientific documents and 2 321 633 inferred from molecular data. This resource aims to provide researchers and clinicians with a centralized repository for potential FDI information that is free and easy to use. Database URL: https://zenodo.org/records/8192515 Database DOI: https://doi.org/10.5281/zenodo.6638469.
Collapse
Affiliation(s)
| | - Oscar Piette
- Computational Biology Group, Precision Nutrition and Cancer Research Program, IMDEA Food Institute, CEI UAM+CSIC, Carretera de Cantoblanco, 8, Madrid 28049, Spain
| | - Marco Garranzo
- Computational Biology Group, Precision Nutrition and Cancer Research Program, IMDEA Food Institute, CEI UAM+CSIC, Carretera de Cantoblanco, 8, Madrid 28049, Spain
| | - David Pérez-Serrano
- Computational Biology Group, Precision Nutrition and Cancer Research Program, IMDEA Food Institute, CEI UAM+CSIC, Carretera de Cantoblanco, 8, Madrid 28049, Spain
| | - Jelena Milešević
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, National Institute of the Republic of Serbia, Tadeuša Košćuška 1, PAK 104 201, Belgrade 11 158, Serbia
- Capacity Development in Nutrition—CAPNUTRA, Trnska 3, Belgrade 11000, Serbia
| | - Isabel Espinosa-Salinas
- GENYAL Platform on Nutrition and Health, IMDEA Food Institute, CEI UAM+CSIC, Carretera de Cantoblanco, 8, Madrid 28049, Spain
| | - Ana Ramírez de Molina
- GENYAL Platform on Nutrition and Health, IMDEA Food Institute, CEI UAM+CSIC, Carretera de Cantoblanco, 8, Madrid 28049, Spain
| | - Teresa Laguna
- Computational Biology Group, Precision Nutrition and Cancer Research Program, IMDEA Food Institute, CEI UAM+CSIC, Carretera de Cantoblanco, 8, Madrid 28049, Spain
| | - Enrique Carrillo de Santa Pau
- Computational Biology Group, Precision Nutrition and Cancer Research Program, IMDEA Food Institute, CEI UAM+CSIC, Carretera de Cantoblanco, 8, Madrid 28049, Spain
| |
Collapse
|
26
|
Muniyappan S, Rayan AXA, Varrieth GT. EGeRepDR: An enhanced genetic-based representation learning for drug repurposing using multiple biomedical sources. J Biomed Inform 2023; 147:104528. [PMID: 37858852 DOI: 10.1016/j.jbi.2023.104528] [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: 07/13/2023] [Revised: 09/11/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
MOTIVATION Drug repurposing (DR) is an imminent approach for identifying novel therapeutic indications for the available drugs and discovering novel drugs for previously untreatable diseases. Nowadays, DR has major attention in the pharmaceutical industry due to the high cost and time of launching new drugs to the market through traditional drug development. DR task majorly depends on genetic information since the drugs revert the modified Gene Expression (GE) of diseases to normal. Many of the existing studies have not considered the genetic importance of predicting the potential candidates. METHOD We proposed a novel multimodal framework that utilizes genetic aspects of drugs and diseases such as genes, pathways, gene signatures, or expression to enhance the performance of DR using various data sources. Firstly, the heterogeneous biological network (HBN) is constructed with three types of nodes namely drug, disease, and gene, and 4 types of edges similarities (drug, gene, and disease), drug-gene, gene-disease, and drug-disease. Next, a modified graph auto-encoder (GAE*) model is applied to learn the representation of drug and disease nodes using the topological structure and edge information. Secondly, the HBN is enhanced with the information extracted from biomedical literature and ontology using a novel semi-supervised pattern embedding-based bootstrapping model and novel DR perspective representation learning respectively to improve the prediction performance. Finally, our proposed system uses a neural network model to generate the probability score of drug-disease pairs. RESULTS We demonstrate the efficiency of the proposed model on various datasets and achieved outstanding performance in 5-fold cross-validation (AUC = 0.99, AUPR = 0.98). Further, we validated the top-ranked potential candidates using pathway analysis and proved that the known and predicted candidates share common genes in the pathways.
Collapse
Affiliation(s)
- Saranya Muniyappan
- Computer Science and Engineering, CEG Campus, Anna University, Chennai, Tamil Nadu, India.
| | | | | |
Collapse
|
27
|
Nguyen TM, Craig DB, Tran D, Nguyen T, Draghici S. A novel approach for predicting upstream regulators (PURE) that affect gene expression. Sci Rep 2023; 13:18571. [PMID: 37903768 PMCID: PMC10616115 DOI: 10.1038/s41598-023-41374-0] [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: 02/06/2023] [Accepted: 08/25/2023] [Indexed: 11/01/2023] Open
Abstract
External factors such as exposure to a chemical, drug, or toxicant (CDT), or conversely, the lack of certain chemicals can cause many diseases. The ability to identify such causal CDTs based on changes in the gene expression profile is extremely important in many studies. Furthermore, the ability to correctly infer CDTs that can revert the gene expression changes induced by a given disease phenotype is a crucial step in drug repurposing. We present an approach for Predicting Upstream REgulators (PURE) designed to tackle this challenge. PURE can correctly infer a CDT from the measured expression changes in a given phenotype, as well as correctly identify drugs that could revert disease-induced gene expression changes. We compared the proposed approach with four classical approaches as well as with the causal analysis used in Ingenuity Pathway Analysis (IPA) on 16 data sets (1 rat, 5 mouse, and 10 human data sets), involving 8 chemicals or drugs. We assessed the results based on the ability to correctly identify the CDT as indicated by its rank. We also considered the number of false positives, i.e. CDTs other than the correct CDT that were reported to be significant by each method. The proposed approach performed best in 11 out of the 16 experiments, reporting the correct CDT at the very top 7 times. IPA was the second best, reporting the correct CDT at the top 5 times, but was unable to identify the correct CDT at all in 5 out of the 16 experiments. The validation results showed that our approach, PURE, outperformed some of the most popular methods in the field. PURE could effectively infer the true CDTs responsible for the observed gene expression changes and could also be useful in drug repurposing applications.
Collapse
Affiliation(s)
- Tuan-Minh Nguyen
- Department of Computer Science, Wayne State University, Detroit, 48202, USA
| | - Douglas B Craig
- Department of Computer Science, Wayne State University, Detroit, 48202, USA
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Duc Tran
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tin Nguyen
- Department of Computer Science and Software Engineering, Auburn University, Auburn, 36849, USA
| | - Sorin Draghici
- Department of Computer Science, Wayne State University, Detroit, 48202, USA.
- Advaita Bioinformatics, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
28
|
He C, Xu Y, Zhou Y, Fan J, Cheng C, Meng R, Gamazon ER, Zhou D. Integrating population-level and cell-based signatures for drug repositioning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.25.564079. [PMID: 37961219 PMCID: PMC10634827 DOI: 10.1101/2023.10.25.564079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Drug repositioning presents a streamlined and cost-efficient way to expand the range of therapeutic possibilities. Furthermore, drugs with genetic evidence are more likely to progress successfully through clinical trials towards FDA approval. Exploiting these developments, single gene-based drug repositioning methods have been implemented, but approaches leveraging the entire spectrum of molecular signatures are critically underexplored. Most multi-gene-based approaches rely on differential gene expression (DGE) analysis, which is prone to identify the molecular consequence of disease and renders causal inference challenging. We propose a framework TReD (Transcriptome-informed Reversal Distance) that integrates population-level disease signatures robust to reverse causality and cell-based drug-induced transcriptome response profiles. TReD embeds the disease signature and drug profile in a high-dimensional normed space, quantifying the reversal potential of candidate drugs in a disease-related cell screen assay. The robustness is ensured by evaluation in additional cell screens. For an application, we implement the framework to identify potential drugs against COVID-19. Taking transcriptome-wide association study (TWAS) results from four relevant tissues and three DGE results as disease features, we identify 37 drugs showing potential reversal roles in at least four of the seven disease signatures. Notably, over 70% (27/37) of the drugs have been linked to COVID-19 from other studies, and among them, eight drugs are supported by ongoing/completed clinical trials. For example, TReD identifies the well-studied JAK1/JAK2 inhibitor baricitinib, the first FDA-approved immunomodulatory treatment for COVID-19. Novel potential candidates, including enzastaurin, a selective inhibitor of PKC-beta which can be activated by SARS-CoV-2, are also identified. In summary, we propose a comprehensive genetics-anchored framework integrating population-level signatures and cell-based screens that can accelerate the search for new therapeutic strategies.
Collapse
|
29
|
Macaya I, Roman M, Welch C, Entrialgo-Cadierno R, Salmon M, Santos A, Feliu I, Kovalski J, Lopez I, Rodriguez-Remirez M, Palomino-Echeverria S, Lonfgren SM, Ferrero M, Calabuig S, Ludwig IA, Lara-Astiaso D, Jantus-Lewintre E, Guruceaga E, Narayanan S, Ponz-Sarvise M, Pineda-Lucena A, Lecanda F, Ruggero D, Khatri P, Santamaria E, Fernandez-Irigoyen J, Ferrer I, Paz-Ares L, Drosten M, Barbacid M, Gil-Bazo I, Vicent S. Signature-driven repurposing of Midostaurin for combination with MEK1/2 and KRASG12C inhibitors in lung cancer. Nat Commun 2023; 14:6332. [PMID: 37816716 PMCID: PMC10564741 DOI: 10.1038/s41467-023-41828-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 09/20/2023] [Indexed: 10/12/2023] Open
Abstract
Drug combinations are key to circumvent resistance mechanisms compromising response to single anti-cancer targeted therapies. The implementation of combinatorial approaches involving MEK1/2 or KRASG12C inhibitors in the context of KRAS-mutated lung cancers focuses fundamentally on targeting KRAS proximal activators or effectors. However, the antitumor effect is highly determined by compensatory mechanisms arising in defined cell types or tumor subgroups. A potential strategy to find drug combinations targeting a larger fraction of KRAS-mutated lung cancers may capitalize on the common, distal gene expression output elicited by oncogenic KRAS. By integrating a signature-driven drug repurposing approach with a pairwise pharmacological screen, here we show synergistic drug combinations consisting of multi-tyrosine kinase PKC inhibitors together with MEK1/2 or KRASG12C inhibitors. Such combinations elicit a cytotoxic response in both in vitro and in vivo models, which in part involves inhibition of the PKC inhibitor target AURKB. Proteome profiling links dysregulation of MYC expression to the effect of both PKC inhibitor-based drug combinations. Furthermore, MYC overexpression appears as a resistance mechanism to MEK1/2 and KRASG12C inhibitors. Our study provides a rational framework for selecting drugs entering combinatorial strategies and unveils MEK1/2- and KRASG12C-based therapies for lung cancer.
Collapse
Affiliation(s)
- Irati Macaya
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
| | - Marta Roman
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Connor Welch
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
| | | | - Marina Salmon
- Experimental Oncology Group, Molecular Oncology Program, Spanish National Cancer Center (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Alba Santos
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Hospital 12 de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Iker Feliu
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
| | - Joanna Kovalski
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
| | - Ines Lopez
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
| | - Maria Rodriguez-Remirez
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
| | - Sara Palomino-Echeverria
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra, Pamplona, Spain
| | - Shane M Lonfgren
- Stanford Institute for Immunity, Transplantation and Infection, Stanford, CA, USA
- Stanford Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Macarena Ferrero
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Molecular Oncology Laboratory, Fundación Para La Investigación del Hospital General Universitario de Valencia, Valencia, Spain
- Mixed Unit TRIAL (Principe Felipe Research Centre & Fundación para la Investigación del Hospital General Universitario de Valencia), Valencia, Spain
| | - Silvia Calabuig
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Molecular Oncology Laboratory, Fundación Para La Investigación del Hospital General Universitario de Valencia, Valencia, Spain
- Mixed Unit TRIAL (Principe Felipe Research Centre & Fundación para la Investigación del Hospital General Universitario de Valencia), Valencia, Spain
- Department of Pathology, Universitat de Valencia, Valencia, Spain
| | - Iziar A Ludwig
- University of Navarra, Center for Applied Medical Research, Molecular Therapies Program, Pamplona, Spain
| | - David Lara-Astiaso
- University of Navarra, Center for Applied Medical Research, Genomics Platform, Pamplona, Spain
| | - Eloisa Jantus-Lewintre
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Molecular Oncology Laboratory, Fundación Para La Investigación del Hospital General Universitario de Valencia, Valencia, Spain
- Mixed Unit TRIAL (Principe Felipe Research Centre & Fundación para la Investigación del Hospital General Universitario de Valencia), Valencia, Spain
- Department of Pathology, Universitat de Valencia, Valencia, Spain
| | - Elizabeth Guruceaga
- University of Navarra, Center for Applied Medical Research, Bioinformatics Platform, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- ProteoRed-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Shruthi Narayanan
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
- Clinica Universidad de Navarra, Department of Medical Oncology, Pamplona, Spain
| | - Mariano Ponz-Sarvise
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Clinica Universidad de Navarra, Department of Medical Oncology, Pamplona, Spain
| | - Antonio Pineda-Lucena
- University of Navarra, Center for Applied Medical Research, Molecular Therapies Program, Pamplona, Spain
| | - Fernando Lecanda
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- University of Navarra, Department of Pathology, Anatomy and Physiology, Pamplona, Spain
| | - Davide Ruggero
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Purvesh Khatri
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra, Pamplona, Spain
| | - Enrique Santamaria
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- ProteoRed-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Joaquin Fernandez-Irigoyen
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- ProteoRed-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Irene Ferrer
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Hospital 12 de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Luis Paz-Ares
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Hospital 12 de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Medical School, Universidad Complutense, Madrid, Spain
| | - Matthias Drosten
- Experimental Oncology Group, Molecular Oncology Program, Spanish National Cancer Center (CNIO), Madrid, Spain
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain
| | - Mariano Barbacid
- Experimental Oncology Group, Molecular Oncology Program, Spanish National Cancer Center (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Gil-Bazo
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Clinica Universidad de Navarra, Department of Medical Oncology, Pamplona, Spain
- Department of Oncology, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Silve Vicent
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
- University of Navarra, Department of Pathology, Anatomy and Physiology, Pamplona, Spain.
| |
Collapse
|
30
|
Li X, Liao M, Wang B, Zan X, Huo Y, Liu Y, Bao Z, Xu P, Liu W. A drug repurposing method based on inhibition effect on gene regulatory network. Comput Struct Biotechnol J 2023; 21:4446-4455. [PMID: 37731599 PMCID: PMC10507583 DOI: 10.1016/j.csbj.2023.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
Numerous computational drug repurposing methods have emerged as efficient alternatives to costly and time-consuming traditional drug discovery approaches. Some of these methods are based on the assumption that the candidate drug should have a reversal effect on disease-associated genes. However, such methods are not applicable in the case that there is limited overlap between disease-related genes and drug-perturbed genes. In this study, we proposed a novel Drug Repurposing method based on the Inhibition Effect on gene regulatory network (DRIE) to identify potential drugs for cancer treatment. DRIE integrated gene expression profile and gene regulatory network to calculate inhibition score by using the shortest path in the disease-specific network. The results on eleven datasets indicated the superior performance of DRIE when compared to other state-of-the-art methods. Case studies showed that our method effectively discovered novel drug-disease associations. Our findings demonstrated that the top-ranked drug candidates had been already validated by CTD database. Additionally, it clearly identified potential agents for three cancers (colorectal, breast, and lung cancer), which was beneficial when annotating drug-disease relationships in the CTD. This study proposed a novel framework for drug repurposing, which would be helpful for drug discovery and development.
Collapse
Affiliation(s)
- Xianbin Li
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, China
| | - Minzhen Liao
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Bing Wang
- School of Medicine, Southeast University, Nanjing, China
| | - Xiangzhen Zan
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Yanhao Huo
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Yue Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Zhenshen Bao
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, China
| | - Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, China
| | - Wenbin Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| |
Collapse
|
31
|
Pividori M, Lu S, Li B, Su C, Johnson ME, Wei WQ, Feng Q, Namjou B, Kiryluk K, Kullo IJ, Luo Y, Sullivan BD, Voight BF, Skarke C, Ritchie MD, Grant SFA, Greene CS. Projecting genetic associations through gene expression patterns highlights disease etiology and drug mechanisms. Nat Commun 2023; 14:5562. [PMID: 37689782 PMCID: PMC10492839 DOI: 10.1038/s41467-023-41057-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 08/18/2023] [Indexed: 09/11/2023] Open
Abstract
Genes act in concert with each other in specific contexts to perform their functions. Determining how these genes influence complex traits requires a mechanistic understanding of expression regulation across different conditions. It has been shown that this insight is critical for developing new therapies. Transcriptome-wide association studies have helped uncover the role of individual genes in disease-relevant mechanisms. However, modern models of the architecture of complex traits predict that gene-gene interactions play a crucial role in disease origin and progression. Here we introduce PhenoPLIER, a computational approach that maps gene-trait associations and pharmacological perturbation data into a common latent representation for a joint analysis. This representation is based on modules of genes with similar expression patterns across the same conditions. We observe that diseases are significantly associated with gene modules expressed in relevant cell types, and our approach is accurate in predicting known drug-disease pairs and inferring mechanisms of action. Furthermore, using a CRISPR screen to analyze lipid regulation, we find that functionally important players lack associations but are prioritized in trait-associated modules by PhenoPLIER. By incorporating groups of co-expressed genes, PhenoPLIER can contextualize genetic associations and reveal potential targets missed by single-gene strategies.
Collapse
Affiliation(s)
- Milton Pividori
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Sumei Lu
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Binglan Li
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA
| | - Chun Su
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Matthew E Johnson
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Wei-Qi Wei
- Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Qiping Feng
- Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Bahram Namjou
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Krzysztof Kiryluk
- Department of Medicine, Division of Nephrology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | | | - Yuan Luo
- Northwestern University, Chicago, IL, 60611, USA
| | - Blair D Sullivan
- Kahlert School of Computing, University of Utah, Salt Lake City, UT, 84112, USA
| | - Benjamin F Voight
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Carsten Skarke
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Marylyn D Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Struan F A Grant
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Casey S Greene
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
- Center for Health AI, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
32
|
Sun J, Xu M, Ru J, James-Bott A, Xiong D, Wang X, Cribbs AP. Small molecule-mediated targeting of microRNAs for drug discovery: Experiments, computational techniques, and disease implications. Eur J Med Chem 2023; 257:115500. [PMID: 37262996 PMCID: PMC11554572 DOI: 10.1016/j.ejmech.2023.115500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/05/2023] [Accepted: 05/15/2023] [Indexed: 06/03/2023]
Abstract
Small molecules have been providing medical breakthroughs for human diseases for more than a century. Recently, identifying small molecule inhibitors that target microRNAs (miRNAs) has gained importance, despite the challenges posed by labour-intensive screening experiments and the significant efforts required for medicinal chemistry optimization. Numerous experimentally-verified cases have demonstrated the potential of miRNA-targeted small molecule inhibitors for disease treatment. This new approach is grounded in their posttranscriptional regulation of the expression of disease-associated genes. Reversing dysregulated gene expression using this mechanism may help control dysfunctional pathways. Furthermore, the ongoing improvement of algorithms has allowed for the integration of computational strategies built on top of laboratory-based data, facilitating a more precise and rational design and discovery of lead compounds. To complement the use of extensive pharmacogenomics data in prioritising potential drugs, our previous work introduced a computational approach based on only molecular sequences. Moreover, various computational tools for predicting molecular interactions in biological networks using similarity-based inference techniques have been accumulated in established studies. However, there are a limited number of comprehensive reviews covering both computational and experimental drug discovery processes. In this review, we outline a cohesive overview of both biological and computational applications in miRNA-targeted drug discovery, along with their disease implications and clinical significance. Finally, utilizing drug-target interaction (DTIs) data from DrugBank, we showcase the effectiveness of deep learning for obtaining the physicochemical characterization of DTIs.
Collapse
Affiliation(s)
- Jianfeng Sun
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK.
| | - Miaoer Xu
- Department of Biology, Emory University, Atlanta, GA, 30322, USA
| | - Jinlong Ru
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, 85354, Germany
| | - Anna James-Bott
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK
| | - Dapeng Xiong
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA; Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Xia Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Adam P Cribbs
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|
33
|
Giudice LC, Oskotsky TT, Falako S, Opoku‐Anane J, Sirota M. Endometriosis in the era of precision medicine and impact on sexual and reproductive health across the lifespan and in diverse populations. FASEB J 2023; 37:e23130. [PMID: 37641572 PMCID: PMC10503213 DOI: 10.1096/fj.202300907] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/26/2023] [Indexed: 08/31/2023]
Abstract
Endometriosis is a common estrogen-dependent disorder wherein uterine lining tissue (endometrium) is found mainly in the pelvis where it causes inflammation, chronic pelvic pain, pain with intercourse and menses, and infertility. Recent evidence also supports a systemic inflammatory component that underlies associated co-morbidities, e.g., migraines and cardiovascular and autoimmune diseases. Genetics and environment contribute significantly to disease risk, and with the explosion of omics technologies, underlying mechanisms of symptoms are increasingly being elucidated, although novel and effective therapeutics for pain and infertility have lagged behind these advances. Moreover, there are stark disparities in diagnosis, access to care, and treatment among persons of color and transgender/nonbinary identity, socioeconomically disadvantaged populations, and adolescents, and a disturbing low awareness among health care providers, policymakers, and the lay public about endometriosis, which, if left undiagnosed and under-treated can lead to significant fibrosis, infertility, depression, and markedly diminished quality of life. This review summarizes endometriosis epidemiology, compelling evidence for its pathogenesis, mechanisms underlying its pathophysiology in the age of precision medicine, recent biomarker discovery, novel therapeutic approaches, and issues around reproductive justice for marginalized populations with this disorder spanning the past 100 years. As we enter the next revolution in health care and biomedical research, with rich molecular and clinical datasets, single-cell omics, and population-level data, endometriosis is well positioned to benefit from data-driven research leveraging computational and artificial intelligence approaches integrating data and predicting disease risk, diagnosis, response to medical and surgical therapies, and prognosis for recurrence.
Collapse
Affiliation(s)
- Linda C. Giudice
- UCSF Stanford Endometriosis Center for Innovation, Training, and Community Outreach (ENACT)University of California, San FranciscoSan FranciscoCaliforniaUSA
- Center for Reproductive SciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Tomiko T. Oskotsky
- UCSF Stanford Endometriosis Center for Innovation, Training, and Community Outreach (ENACT)University of California, San FranciscoSan FranciscoCaliforniaUSA
- Bakar Computational Health Sciences InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Simileoluwa Falako
- UCSF Stanford Endometriosis Center for Innovation, Training, and Community Outreach (ENACT)University of California, San FranciscoSan FranciscoCaliforniaUSA
- Columbia University Vagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
| | - Jessica Opoku‐Anane
- UCSF Stanford Endometriosis Center for Innovation, Training, and Community Outreach (ENACT)University of California, San FranciscoSan FranciscoCaliforniaUSA
- Division of Gynecologic Specialty SurgeryColumbia UniversityNew YorkNew YorkUSA
| | - Marina Sirota
- UCSF Stanford Endometriosis Center for Innovation, Training, and Community Outreach (ENACT)University of California, San FranciscoSan FranciscoCaliforniaUSA
- Bakar Computational Health Sciences InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of PediatricsUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
34
|
Irham LM, Adikusuma W, La’ah AS, Chong R, Septama AW, Angelina M. Leveraging Genomic and Bioinformatic Analysis to Enhance Drug Repositioning for Dermatomyositis. Bioengineering (Basel) 2023; 10:890. [PMID: 37627776 PMCID: PMC10451728 DOI: 10.3390/bioengineering10080890] [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: 03/02/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 08/27/2023] Open
Abstract
Dermatomyositis (DM) is an autoimmune disease that is classified as a type of idiopathic inflammatory myopathy, which affects human skin and muscles. The most common clinical symptoms of DM are muscle weakness, rash, and scaly skin. There is currently no cure for DM. Genetic factors are known to play a pivotal role in DM progression, but few have utilized this information geared toward drug discovery for the disease. Here, we exploited genomic variation associated with DM and integrated this with genomic and bioinformatic analyses to discover new drug candidates. We first integrated genome-wide association study (GWAS) and phenome-wide association study (PheWAS) catalogs to identify disease-associated genomic variants. Biological risk genes for DM were prioritized using strict functional annotations, further identifying candidate drug targets based on druggable genes from databases. Overall, we analyzed 1239 variants associated with DM and obtained 43 drugs that overlapped with 13 target genes (JAK2, FCGR3B, CD4, CD3D, LCK, CD2, CD3E, FCGR3A, CD3G, IFNAR1, CD247, JAK1, IFNAR2). Six drugs clinically investigated for DM, as well as eight drugs under pre-clinical investigation, are candidate drugs that could be repositioned for DM. Further studies are necessary to validate potential biomarkers for novel DM therapeutics from our findings.
Collapse
Affiliation(s)
- Lalu Muhammad Irham
- Faculty of Pharmacy, Universitas Ahmad Dahlan, Yogyakarta 55164, Indonesia
- Research Centre for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), South Tangerang 15314, Indonesia
| | - Wirawan Adikusuma
- Department of Pharmacy, University of Muhammadiyah Mataram, Mataram 83127, Indonesia
- Research Center for Vaccine and Drugs, National Research and Innovation Agency (BRIN), South Tangerang 15314, Indonesia
| | - Anita Silas La’ah
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 112304, Taiwan
| | - Rockie Chong
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Abdi Wira Septama
- Research Centre for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), South Tangerang 15314, Indonesia
| | - Marissa Angelina
- Research Centre for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), South Tangerang 15314, Indonesia
| |
Collapse
|
35
|
Yu K, Basu A, Yau C, Wolf DM, Goodarzi H, Bandyopadhyay S, Korkola JE, Hirst GL, Asare S, DeMichele A, Hylton N, Yee D, Esserman L, van ‘t Veer L, Sirota M. Computational drug repositioning for the identification of new agents to sensitize drug-resistant breast tumors across treatments and receptor subtypes. Front Oncol 2023; 13:1192208. [PMID: 37384294 PMCID: PMC10294228 DOI: 10.3389/fonc.2023.1192208] [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: 03/23/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction Drug resistance is a major obstacle in cancer treatment and can involve a variety of different factors. Identifying effective therapies for drug resistant tumors is integral for improving patient outcomes. Methods In this study, we applied a computational drug repositioning approach to identify potential agents to sensitize primary drug resistant breast cancers. We extracted drug resistance profiles from the I-SPY 2 TRIAL, a neoadjuvant trial for early stage breast cancer, by comparing gene expression profiles of responder and non-responder patients stratified into treatments within HR/HER2 receptor subtypes, yielding 17 treatment-subtype pairs. We then used a rank-based pattern-matching strategy to identify compounds in the Connectivity Map, a database of cell line derived drug perturbation profiles, that can reverse these signatures in a breast cancer cell line. We hypothesize that reversing these drug resistance signatures will sensitize tumors to treatment and prolong survival. Results We found that few individual genes are shared among the drug resistance profiles of different agents. At the pathway level, however, we found enrichment of immune pathways in the responders in 8 treatments within the HR+HER2+, HR+HER2-, and HR-HER2- receptor subtypes. We also found enrichment of estrogen response pathways in the non-responders in 10 treatments primarily within the hormone receptor positive subtypes. Although most of our drug predictions are unique to treatment arms and receptor subtypes, our drug repositioning pipeline identified the estrogen receptor antagonist fulvestrant as a compound that can potentially reverse resistance across 13/17 of the treatments and receptor subtypes including HR+ and triple negative. While fulvestrant showed limited efficacy when tested in a panel of 5 paclitaxel resistant breast cancer cell lines, it did increase drug response in combination with paclitaxel in HCC-1937, a triple negative breast cancer cell line. Conclusion We applied a computational drug repurposing approach to identify potential agents to sensitize drug resistant breast cancers in the I-SPY 2 TRIAL. We identified fulvestrant as a potential drug hit and showed that it increased response in a paclitaxel-resistant triple negative breast cancer cell line, HCC-1937, when treated in combination with paclitaxel.
Collapse
Affiliation(s)
- Katharine Yu
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Amrita Basu
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Denise M. Wolf
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Hani Goodarzi
- University of California, San Francisco, San Francisco, CA, United States
| | | | - James E. Korkola
- Oregon Health and Science University, Portland, OR, United States
| | - Gillian L. Hirst
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Smita Asare
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- QuantumLeap Healthcare Collaborative, San Francisco, CA, United States
| | | | - Nola Hylton
- University of California, San Francisco, San Francisco, CA, United States
| | - Douglas Yee
- University of Minnesota, Minneapolis, MN, United States
| | - Laura Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Laura van ‘t Veer
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
36
|
Engler Hart C, Ence D, Healey D, Domingo-Fernández D. On the correspondence between the transcriptomic response of a compound and its effects on its targets. BMC Bioinformatics 2023; 24:207. [PMID: 37208587 DOI: 10.1186/s12859-023-05337-6] [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: 01/30/2023] [Accepted: 05/14/2023] [Indexed: 05/21/2023] Open
Abstract
Better understanding the transcriptomic response produced by a compound perturbing its targets can shed light on the underlying biological processes regulated by the compound. However, establishing the relationship between the induced transcriptomic response and the target of a compound is non-trivial, partly because targets are rarely differentially expressed. Therefore, connecting both modalities requires orthogonal information (e.g., pathway or functional information). Here, we present a comprehensive study aimed at exploring this relationship by leveraging thousands of transcriptomic experiments and target data for over 2000 compounds. Firstly, we confirm that compound-target information does not correlate as expected with the transcriptomic signatures induced by a compound. However, we reveal how the concordance between both modalities increases by connecting pathway and target information. Additionally, we investigate whether compounds that target the same proteins induce a similar transcriptomic response and conversely, whether compounds with similar transcriptomic responses share the same target proteins. While our findings suggest that this is generally not the case, we did observe that compounds with similar transcriptomic profiles are more likely to share at least one protein target and common therapeutic applications. Finally, we demonstrate how to exploit the relationship between both modalities for mechanism of action deconvolution by presenting a case scenario involving a few compound pairs with high similarity.
Collapse
|
37
|
Baglaenko Y, Wagner C, Bhoj VG, Brodin P, Gershwin ME, Graham D, Invernizzi P, Kidd KK, Korsunsky I, Levy M, Mammen AL, Nizet V, Ramirez-Valle F, Stites EC, Williams MS, Wilson M, Rose NR, Ladd V, Sirota M. Making inroads to precision medicine for the treatment of autoimmune diseases: Harnessing genomic studies to better diagnose and treat complex disorders. CAMBRIDGE PRISMS. PRECISION MEDICINE 2023; 1:e25. [PMID: 38550937 PMCID: PMC10953750 DOI: 10.1017/pcm.2023.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 06/13/2024]
Abstract
Precision Medicine is an emerging approach for disease treatment and prevention that takes into account individual variability in genes, environment, and lifestyle. Autoimmune diseases are those in which the body's natural defense system loses discriminating power between its own cells and foreign cells, causing the body to mistakenly attack healthy tissues. These conditions are very heterogeneous in their presentation and therefore difficult to diagnose and treat. Achieving precision medicine in autoimmune diseases has been challenging due to the complex etiologies of these conditions, involving an interplay between genetic, epigenetic, and environmental factors. However, recent technological and computational advances in molecular profiling have helped identify patient subtypes and molecular pathways which can be used to improve diagnostics and therapeutics. This review discusses the current understanding of the disease mechanisms, heterogeneity, and pathogenic autoantigens in autoimmune diseases gained from genomic and transcriptomic studies and highlights how these findings can be applied to better understand disease heterogeneity in the context of disease diagnostics and therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniel Graham
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
| | - Kenneth K. Kidd
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Michael Levy
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew L. Mammen
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, USA
| | - Victor Nizet
- School of Medicine, University of California San Diego, San Diego, CA, USA
| | | | - Edward C. Stites
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Michael Wilson
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, CA, USA
| | - Noel R. Rose
- Autoimmune Association, Clinton Township, MI, USA
| | | | - Marina Sirota
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| |
Collapse
|
38
|
Ahmed F, Samantasinghar A, Manzoor Soomro A, Kim S, Hyun Choi K. A systematic review of computational approaches to understand cancer biology for informed drug repurposing. J Biomed Inform 2023; 142:104373. [PMID: 37120047 DOI: 10.1016/j.jbi.2023.104373] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/25/2023] [Accepted: 04/23/2023] [Indexed: 05/01/2023]
Abstract
Cancer is the second leading cause of death globally, trailing only heart disease. In the United States alone, 1.9 million new cancer cases and 609,360 deaths were recorded for 2022. Unfortunately, the success rate for new cancer drug development remains less than 10%, making the disease particularly challenging. This low success rate is largely attributed to the complex and poorly understood nature of cancer etiology. Therefore, it is critical to find alternative approaches to understanding cancer biology and developing effective treatments. One such approach is drug repurposing, which offers a shorter drug development timeline and lower costs while increasing the likelihood of success. In this review, we provide a comprehensive analysis of computational approaches for understanding cancer biology, including systems biology, multi-omics, and pathway analysis. Additionally, we examine the use of these methods for drug repurposing in cancer, including the databases and tools that are used for cancer research. Finally, we present case studies of drug repurposing, discussing their limitations and offering recommendations for future research in this area.
Collapse
Affiliation(s)
- Faheem Ahmed
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea
| | | | | | - Sejong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea.
| |
Collapse
|
39
|
Yuan Y, Zhang Y, Meng X, Liu Z, Wang B, Miao R, Zhang R, Su W, Liu L. EDC-DTI: An end-to-end deep collaborative learning model based on multiple information for drug-target interactions prediction. J Mol Graph Model 2023; 122:108498. [PMID: 37126908 DOI: 10.1016/j.jmgm.2023.108498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
Innovations in drug-target interactions (DTIs) prediction accelerate the progression of drug development. The introduction of deep learning models has a dramatic impact on DTIs prediction, with a distinct influence on saving time and money in drug discovery. This study develops an end-to-end deep collaborative learning model for DTIs prediction, called EDC-DTI, to identify new targets for existing drugs based on multiple drug-target-related information including homogeneous information and heterogeneous information by the way of deep learning. Our end-to-end model is composed of a feature builder and a classifier. Feature builder consists of two collaborative feature construction algorithms that extract the molecular properties and the topology property of networks, and the classifier consists of a feature encoder and a feature decoder which are designed for feature integration and DTIs prediction, respectively. The feature encoder, mainly based on the improved graph attention network, incorporates heterogeneous information into drug features and target features separately. The feature decoder is composed of multiple neural networks for predictions. Compared with six popular baseline models, EDC-DTI achieves highest predictive performance in the case of low computational costs. Robustness tests demonstrate that EDC-DTI is able to maintain strong predictive performance on sparse datasets. As well, we use the model to predict the most likely targets to interact with Simvastatin (DB00641), Nifedipine (DB01115) and Afatinib (DB08916) as examples. Results show that most of the predictions can be confirmed by literature with clear evidence.
Collapse
Affiliation(s)
- Yongna Yuan
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China.
| | - Yuhao Zhang
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China
| | - Xiangbo Meng
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China
| | - Zhenyu Liu
- School of Cyberspace Security, Gansu University of Political Science and Law, Anning West Road, Lanzhou, 730070, Gansu, China
| | - Bohan Wang
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China
| | - Ruidong Miao
- School of Life Science, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China
| | - Ruisheng Zhang
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China
| | - Wei Su
- School of Information Science & Engineering, Lanzhou University, South Tianshui Road, Lanzhou, 730000, Gansu, China
| | - Lei Liu
- Duzhe Publishing Group Co. Ltd., DuZhe Road, Lanzhou, 730000, Gansu, China
| |
Collapse
|
40
|
Wang Z, Mehmood A, Yao J, Zhang H, Wang L, Al-Shehri M, Kaushik AC, Wei DQ. Combination of furosemide, gold, and dopamine as a potential therapy for breast cancer. Funct Integr Genomics 2023; 23:94. [PMID: 36943579 DOI: 10.1007/s10142-023-01007-1] [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: 07/27/2022] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/23/2023]
Abstract
Breast cancer is one of the leading causes of death in women worldwide. Initially, it develops in the epithelium of the ducts or lobules of the breast glandular tissues with limited growth and the potential to metastasize. It is a highly heterogeneous malignancy; however, the common molecular mechanisms could help identify new targeted drugs for treating its subtypes. This study uses computational drug repositioning approaches to explore fresh drug candidates for breast cancer treatment. We also implemented reversal gene expression and gene expression-based signatures to explore novel drug candidates computationally. The drug activity profiles and related gene expression changes were acquired from the DrugBank, PubChem, and LINCS databases, and then in silico drug screening, molecular dynamics (MD) simulation, replica exchange MD simulations, and simulated annealing molecular dynamics (SAMD) simulations were conducted to discover and verify the valid drug candidates. We have found that compounds like furosemide, gold, and dopamine showed significant outcomes. Furthermore, the expression of genes related to breast cancer was observed to be reversed by these shortlisted drugs. Therefore, we postulate that combining furosemide, gold, and dopamine would be a potential combination therapy measurement for breast cancer patients.
Collapse
Affiliation(s)
- Zhen Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Aamir Mehmood
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jia Yao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hui Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Li Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Mohammed Al-Shehri
- Department of Biology, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | | | - Dong-Qing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Nanyang, Henan, China.
- Peng Cheng Laboratory, Nanshan District, Shenzhen, Guangdong, China.
| |
Collapse
|
41
|
Maria NI, Rapicavoli RV, Alaimo S, Bischof E, Stasuzzo A, Broek JA, Pulvirenti A, Mishra B, Duits AJ, Ferro A. Application of the PHENotype SIMulator for rapid identification of potential candidates in effective COVID-19 drug repurposing. Heliyon 2023; 9:e14115. [PMID: 36911878 PMCID: PMC9986505 DOI: 10.1016/j.heliyon.2023.e14115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
The current, rapidly diversifying pandemic has accelerated the need for efficient and effective identification of potential drug candidates for COVID-19. Knowledge on host-immune response to SARS-CoV-2 infection, however, remains limited with few drugs approved to date. Viable strategies and tools are rapidly arising to address this, especially with repurposing of existing drugs offering significant promise. Here we introduce a systems biology tool, the PHENotype SIMulator, which -by leveraging available transcriptomic and proteomic databases-allows modeling of SARS-CoV-2 infection in host cells in silico to i) determine with high sensitivity and specificity (both>96%) the viral effects on cellular host-immune response, resulting in specific cellular SARS-CoV-2 signatures and ii) utilize these cell-specific signatures to identify promising repurposable therapeutics. Powered by this tool, coupled with domain expertise, we identify several potential COVID-19 drugs including methylprednisolone and metformin, and further discern key cellular SARS-CoV-2-affected pathways as potential druggable targets in COVID-19 pathogenesis.
Collapse
Key Words
- 2DG, 2-Deoxy-Glucose
- ACE2, Angiotensin-converting enzyme 2
- COVID-19
- COVID-19, Coronavirus disease 2019
- Caco-2, Human colon epithelial carcinoma cell line
- Calu-3, Epithelial cell line
- Cellular SARS-CoV-2 signatures
- Cellular host-immune response
- Cellular simulation models
- DEGs, Differentially Expressed Genes
- DEPs, Differentially expressed proteins
- Drug repurposing
- HCQ-CQ, (Hydroxy)chloroquine
- IFN, Interferon
- ISGs, IFN-stimulated genes
- MITHrIL, Mirna enrIched paTHway Impact anaLysis
- MOI, Multiplicity of infection
- MP, Methylprednisolone
- NHBE, Normal human bronchial epithelial cells
- PHENSIM, PHENotype SIMulator
- SARS-CoV-2, Severe acute respiratory syndrome coronavirus 2
- Systems biology
- TLR, Toll-like Receptor
Collapse
Affiliation(s)
- Naomi I. Maria
- Department of Computer Science, Mathematics, Engineering and Cell Biology, Courant Institute, Tandon and School of Medicine, New York University, New York, USA
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra, Northwell Health, Manhasset, NY, USA
- Red Cross Blood Bank Foundation Curaçao, Willemstad, Curaçao
- Department of Medical Microbiology and Immunology, St. Antonius Ziekenhuis, Niewegein, the Netherlands
- Corresponding author. Department of Computer Science, Mathematics, Engineering and Cell Biology, Courant Institute, Tandon and School of Medicine, New York University, New York, USA.
| | - Rosaria Valentina Rapicavoli
- Department of Physics and Astronomy, University of Catania, Italy
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - Salvatore Alaimo
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - Evelyne Bischof
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, Naples, Italy
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Pudong, Shanghai, China
- Insilico Medicine, Hong Kong Special Administrative Region, China
| | | | - Jantine A.C. Broek
- Department of Computer Science, Mathematics, Engineering and Cell Biology, Courant Institute, Tandon and School of Medicine, New York University, New York, USA
| | - Alfredo Pulvirenti
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | - Bud Mishra
- Department of Computer Science, Mathematics, Engineering and Cell Biology, Courant Institute, Tandon and School of Medicine, New York University, New York, USA
- Simon Center for Quantitative Biology, Cold Spring Harbor Lab, Long Island, USA
- Corresponding author. Courant Institute of Mathematical Sciences, Room 405, 251 Mercer Street, NY, USA.
| | - Ashley J. Duits
- Red Cross Blood Bank Foundation Curaçao, Willemstad, Curaçao
- Curaçao Biomedical Health Research Institute, Willemstad, Curaçao
- Institute for Medical Education, University Medical Center Groningen, Groningen, the Netherlands
| | - Alfredo Ferro
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, Italy
| | | |
Collapse
|
42
|
Miura S, Sawada R, Yorita A, Kida H, Kamada T, Yamanishi Y. A trial of topiramate for patients with hereditary spinocerebellar ataxia. Clin Case Rep 2023; 11:e6980. [PMID: 36855409 PMCID: PMC9968455 DOI: 10.1002/ccr3.6980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/24/2022] [Accepted: 02/06/2023] [Indexed: 02/27/2023] Open
Abstract
In an open pilot trial, six patients with various hereditary forms of spinocerebellar ataxia (SCA) were assigned to topiramate (50 mg/day) for 24 weeks. Four patients completed the protocol without adverse events. Of these four patients, topiramate was effective for three patients. Some patients with SCA could respond to treatment with topiramate.
Collapse
Affiliation(s)
- Shiroh Miura
- Department of Neurology and Geriatric MedicineEhime University Graduate School of MedicineToonEhimeJapan
| | - Ryusuke Sawada
- Department of PharmacologyOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesKita‐kuOkayamaJapan
| | - Akiko Yorita
- Division of Respirology, Neurology and Rheumatology, Department of MedicineKurume University School of MedicineKurumeFukuokaJapan
| | - Hiroshi Kida
- Department of AnatomyFukuoka University School of MedicineJonan‐kuFukuokaJapan
| | - Takashi Kamada
- Department of NeurologyFukuoka Sanno HospitalSawara‐kuFukuokaJapan
| | - Yoshihiro Yamanishi
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems EngineeringKyushu Institute of TechnologyIizukaFukuokaJapan
| |
Collapse
|
43
|
In vivo near-infrared fluorescence and SPECT-CT imaging of colorectal Cancer using the bradykinin B2R-specific ligand icatibant. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 239:112648. [PMID: 36641883 DOI: 10.1016/j.jphotobiol.2023.112648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/25/2022] [Accepted: 01/08/2023] [Indexed: 01/11/2023]
Abstract
Cancer molecular imaging using specific probes designed to identify target proteins in cancer is a powerful tool to guide therapeutic selection, patient management, and follow-up. We demonstrated that icatibant may be used as a targeting probe for the significantly upregulated bradykinin B2R in colorectal cancer (CRC). Icatibant-based probes with high affinity towards bradykinin B2R were identified. The near-infrared (NIR) fluorescent dye conjugate MPA-PEG3-k-Icatibant and radioconjugate [99mTc]Tc-HYNIC-PEG4-Icatibant exhibited favourable selective and specific uptake in tumours when the subcutaneous and orthotopic colorectal tumour-bearing mouse models were imaged using NIR fluorescence imaging and Single-Photon Emission Computed Tomography-Computed Tomography (SPECT-CT), respectively. The tracer of [99mTc]Tc-HYNIC-PEG4-Icatibant accumulated in tumours according to biodistribution studies and peaked at 4 h with an uptake value of 3.41 ± 0.27%ID/g in HT29 tumour-bearing nude mice following intravenous injection (i.v.). The tumour-to-colorectal signal ratios were 5.03 ± 0.37, 15.45 ± 0.32, 13.58 ± 1.19 and 11.33 ± 1.73 1, 2, 4 and 6 h after tail-veil injection, respectively. Overall, in the wake of rapid and precise tumour delineation and penetration characteristics, icatibant-based probes represent promising high-contrast molecular imaging probes for the detection of bradykinin B2R.
Collapse
|
44
|
Hsieh KL, Plascencia-Villa G, Lin KH, Perry G, Jiang X, Kim Y. Synthesize heterogeneous biological knowledge via representation learning for Alzheimer's disease drug repurposing. iScience 2023; 26:105678. [PMID: 36594024 PMCID: PMC9804117 DOI: 10.1016/j.isci.2022.105678] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/04/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Developing drugs for treating Alzheimer's disease has been extremely challenging and costly due to limited knowledge of underlying mechanisms and therapeutic targets. To address the challenge in AD drug development, we developed a multi-task deep learning pipeline that learns biological interactions and AD risk genes, then utilizes multi-level evidence on drug efficacy to identify repurposable drug candidates. Using the embedding derived from the model, we ranked drug candidates based on evidence from post-treatment transcriptomic patterns, efficacy in preclinical models, population-based treatment effects, and clinical trials. We mechanistically validated the top-ranked candidates in neuronal cells, identifying drug combinations with efficacy in reducing oxidative stress and safety in maintaining neuronal viability and morphology. Our neuronal response experiments confirmed several biologically efficacious drug combinations. This pipeline showed that harmonizing heterogeneous and complementary data/knowledge, including human interactome, transcriptome patterns, experimental efficacy, and real-world patient data shed light on the drug development of complex diseases.
Collapse
Affiliation(s)
- Kang-Lin Hsieh
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - German Plascencia-Villa
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX 78729, USA
| | - Ko-Hong Lin
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX 78729, USA
| | - Xiaoqian Jiang
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yejin Kim
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
45
|
Sun J, Ru J, Ramos-Mucci L, Qi F, Chen Z, Chen S, Cribbs AP, Deng L, Wang X. DeepsmirUD: Prediction of Regulatory Effects on microRNA Expression Mediated by Small Molecules Using Deep Learning. Int J Mol Sci 2023; 24:1878. [PMID: 36768205 PMCID: PMC9915273 DOI: 10.3390/ijms24031878] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/26/2022] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
Aberrant miRNA expression has been associated with a large number of human diseases. Therefore, targeting miRNAs to regulate their expression levels has become an important therapy against diseases that stem from the dysfunction of pathways regulated by miRNAs. In recent years, small molecules have demonstrated enormous potential as drugs to regulate miRNA expression (i.e., SM-miR). A clear understanding of the mechanism of action of small molecules on the upregulation and downregulation of miRNA expression allows precise diagnosis and treatment of oncogenic pathways. However, outside of a slow and costly process of experimental determination, computational strategies to assist this on an ad hoc basis have yet to be formulated. In this work, we developed, to the best of our knowledge, the first cross-platform prediction tool, DeepsmirUD, to infer small-molecule-mediated regulatory effects on miRNA expression (i.e., upregulation or downregulation). This method is powered by 12 cutting-edge deep-learning frameworks and achieved AUC values of 0.843/0.984 and AUCPR values of 0.866/0.992 on two independent test datasets. With a complementarily constructed network inference approach based on similarity, we report a significantly improved accuracy of 0.813 in determining the regulatory effects of nearly 650 associated SM-miR relations, each formed with either novel small molecule or novel miRNA. By further integrating miRNA-cancer relationships, we established a database of potential pharmaceutical drugs from 1343 small molecules for 107 cancer diseases to understand the drug mechanisms of action and offer novel insight into drug repositioning. Furthermore, we have employed DeepsmirUD to predict the regulatory effects of a large number of high-confidence associated SM-miR relations. Taken together, our method shows promise to accelerate the development of potential miRNA targets and small molecule drugs.
Collapse
Affiliation(s)
- Jianfeng Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Jinlong Ru
- Institute of Virology, Helmholtz Centre Munich—German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Lorenzo Ramos-Mucci
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Fei Qi
- Institute of Genomics, School of Medicine, Huaqiao University, Xiamen 362021, China
| | - Zihao Chen
- Department of Computational Biology for Drug Discovery, Biolife Biotechnology Ltd., Zhumadian 463200, China
| | - Suyuan Chen
- Leibniz-Institut für Analytische Wissenschaften–ISAS–e.V., Otto-Hahn-Str asse 6b, 44227 Dortmund, Germany
| | - Adam P. Cribbs
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Li Deng
- Institute of Virology, Helmholtz Centre Munich—German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Chair of Prevention of Microbial Diseases, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Xia Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
46
|
Liang XZ, Liu XC, Li S, Wen MT, Chen YR, Luo D, Xu B, Li NH, Li G. IRF8 and its related molecules as potential diagnostic biomarkers or therapeutic candidates and immune cell infiltration characteristics in steroid-induced osteonecrosis of the femoral head. J Orthop Surg Res 2023; 18:27. [PMID: 36627660 PMCID: PMC9832881 DOI: 10.1186/s13018-022-03381-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 11/02/2022] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Steroid-induced osteonecrosis of the femoral head (SONFH) was a refractory orthopedic hip joint disease in the young and middle-aged people, but the pathogenesis of SONFH remained unclear. We aimed to identify the potential genes and screen potential therapeutic compounds for SONFH. METHODS The microarray was obtained for blood tissue from the GEO database, and then it identifies differentially expressed genes (DEGs). The DEGs were analyzed to obtain the differences in immune cell infiltration. The gene functional enrichment analysis of SONFH was analyzed. The PPI of DEGs was identified through the STRING database, and the cluster modules and hub genes were ascertained using MCODE and CytoHubba, and the ROC curve of hub genes was analyzed, and the tissues distribution of hub genes was understood by the HPA, Bgee and BioGPS databases. The hub genes and target miRNAs and corresponding upstream lncRNAs were predicted by TargetScan, miRDB and ENCORI database. Subsequently, we used CMap, DGIdb and L1000FWD databases to identify several potential therapeutic molecular compounds for SONFH. Finally, the AutoDockTools Vina, PyMOL and Discovery Studio were employed for molecular docking analyses between compounds and hub genes. RESULTS The microarray dataset GSE123568 was obtained related to SONFH. There were 372 DEGs including 197 upregulated genes and 175 downregulated genes by adjusted P value < 0.01 and |log2FC|> 1. Several significant GSEA enrichment analysis and biological processes and KEGG pathway associated with SONFH were identified, which were significantly related to cytoskeleton organization, nucleobase-containing compound catabolic process, NOD-like receptor signaling pathway, MAPK signaling pathway, FoxO signaling pathway, neutrophil-mediated immunity, neutrophil degranulation and neutrophil activation involved in immune response. Activated T cells CD4 memory, B cells naïve, B cells memory, T cells CD8 and T cells gamma delta might be involved in the occurrence and development of SONFH. Three cluster modules were identified in the PPI network, and eleven hub genes including FPR2, LILRB2, MNDA, CCR1, IRF8, TYROBP, TLR1, HCK, TLR8, TLR2 and CCR2 were identified by Cytohubba, which were differed in bone marrow, adipose tissue and blood, and which had good diagnostic performance in SONFH. We identified IRF8 and 10 target miRNAs that was utilized including Targetsan, miRDB and ENCORI databases and 8 corresponding upstream lncRNAs that was revealed by ENCORI database. IRF8 was detected with consistent expression by qRT-PCR. Based on the CMap, DGIdb and L1000FWD databases, the 11 small molecular compounds that were most strongly therapeutic correlated with SONFH were estradiol, genistein, domperidone, lovastatin, myricetin, fenbufen, rosiglitazone, sirolimus, phenformin, vorinostat and vinblastine. All of 11 small molecules had good binding affinity with the IRF8 in molecular docking. CONCLUSION The occurrence of SONFH was associated with a "multi-target" and "multi-pathway" pattern, especially related to immunity, and IRF8 and its noncoding RNA were closely related to the development of SONFH. The CMap, DGIdb and L1000FWD databases could be effectively used in a systematic manner to predict potential drugs for the prevention and treatment of SONFH. However, additional clinical and experimental research is warranted.
Collapse
Affiliation(s)
- Xue-Zhen Liang
- Orthopaedic Microsurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Jinan City, 250014 Shandong Province China
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355 Shandong China
| | - Xiao-Chen Liu
- Orthopaedic Microsurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Jinan City, 250014 Shandong Province China
| | - Song Li
- Library, Shandong University of Traditional Chinese Medicine, Jinan, 250355 Shandong China
| | - Ming-Tao Wen
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355 Shandong China
| | - Yan-Rong Chen
- Orthopaedic Microsurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Jinan City, 250014 Shandong Province China
| | - Di Luo
- Orthopaedic Microsurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Jinan City, 250014 Shandong Province China
| | - Bo Xu
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355 Shandong China
| | - Nian-Hu Li
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355 Shandong China
- Spinal Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Jinan City, 250014 Shandong Province China
| | - Gang Li
- Orthopaedic Microsurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Jinan City, 250014 Shandong Province China
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355 Shandong China
| |
Collapse
|
47
|
Doumat G, Daher D, Zerdan MB, Nasra N, Bahmad HF, Recine M, Poppiti R. Drug Repurposing in Non-Small Cell Lung Carcinoma: Old Solutions for New Problems. Curr Oncol 2023; 30:704-719. [PMID: 36661704 PMCID: PMC9858415 DOI: 10.3390/curroncol30010055] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Lung cancer is the second most common cancer and the leading cause of cancer-related deaths in 2022. The majority (80%) of lung cancer cases belong to the non-small cell lung carcinoma (NSCLC) subtype. Despite the increased screening efforts, the median five-year survival of metastatic NSCLC remains low at approximately 3%. Common treatment approaches for NSCLC include surgery, multimodal chemotherapy, and concurrent radio and chemotherapy. NSCLC exhibits high rates of resistance to treatment, driven by its heterogeneity and the plasticity of cancer stem cells (CSCs). Drug repurposing offers a faster and cheaper way to develop new antineoplastic purposes for existing drugs, to help overcome therapy resistance. The decrease in time and funds needed stems from the availability of the pharmacokinetic and pharmacodynamic profiles of the Food and Drug Administration (FDA)-approved drugs to be repurposed. This review provides a synopsis of the drug-repurposing approaches and mechanisms of action of potential candidate drugs used in treating NSCLC, including but not limited to antihypertensives, anti-hyperlipidemics, anti-inflammatory drugs, anti-diabetics, and anti-microbials.
Collapse
Affiliation(s)
- George Doumat
- Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Darine Daher
- Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Morgan Bou Zerdan
- Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Nasri Nasra
- Faculty of Medicine, University of Aleppo, Aleppo 15310, Syria
| | - Hisham F. Bahmad
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | - Monica Recine
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Robert Poppiti
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
48
|
Drug-disease association prediction based on end-to-end multi-layer heterogeneous graph convolutional encoders. INFORMATICS IN MEDICINE UNLOCKED 2023. [DOI: 10.1016/j.imu.2023.101177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
49
|
Mondal AK, Swaroop A. Network Biology and Medicine to Rescue: Applications for Retinal Disease Mechanisms and Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:165-171. [PMID: 37440030 PMCID: PMC11377069 DOI: 10.1007/978-3-031-27681-1_25] [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] [Indexed: 07/14/2023]
Abstract
Inherited retinal degenerations (IRDs) are clinically and genetically heterogenous blinding diseases that manifest through dysfunction of target cells, photoreceptors, and retinal pigment epithelium (RPE) in the retina. Despite knowledge of numerous underlying genetic defects, current therapeutic approaches, including gene centric applications, have had limited success, thereby asserting the need of new directions for basic and translational research. Human diseases have commonalities that can be represented in a network form, called diseasome, which captures relationships among disease genes, proteins, metabolites, and patient meta-data. Clinical and genetic information of IRDs suggest shared relationships among pathobiological factors, making these a model case for network medicine. Characterization of the diseasome would considerably improve our understanding of retinal pathologies and permit better design of targeted therapies for disrupted regions within the integrated disease network. Network medicine in synergy with the ongoing artificial intelligence revolution can boost therapeutic developments, especially gene agnostic treatment opportunities.
Collapse
Affiliation(s)
- Anupam K Mondal
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Anand Swaroop
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
50
|
Lang X, Liu J, Zhang G, Feng X, Dan W. Knowledge Mapping of Drug Repositioning's Theme and Development. Drug Des Devel Ther 2023; 17:1157-1174. [PMID: 37096060 PMCID: PMC10122475 DOI: 10.2147/dddt.s405906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/11/2023] [Indexed: 04/26/2023] Open
Abstract
Background In recent years, the emergence of new diseases and resistance to known diseases have led to increasing demand for new drugs. By means of bibliometric analysis, this paper studied the relevant articles on drug repositioning in recent years and analyzed the current research foci and trends. Methodology The Web of Science database was searched to collect all relevant literature on drug repositioning from 2001 to 2022. These data were imported into CiteSpace and bibliometric online analysis platforms for bibliometric analysis. The processed data and visualized images predict the development trends in the research field. Results The quality and quantity of articles published after 2011 have improved significantly, with 45 of them cited more than 100 times. Articles posted by journals from different countries have high citation values. Authors from other institutions have also collaborated to analyze drug rediscovery. Keywords found in the literature include molecular docking (N=223), virtual screening (N=170), drug discovery (N=126), machine learning (N=125), and drug-target interaction (N=68); these words represent the core content of drug repositioning. Conclusion The key focus of drug research and development is related to the discovery of new indications for drugs. Researchers are starting to retarget drugs after analyzing online databases and clinical trials. More and more drugs are being targeted at other diseases to treat more patients, based on saving money and time. It is worth noting that researchers need more financial and technical support to complete drug development.
Collapse
Affiliation(s)
- Xiaona Lang
- Pharmacy Department, Tianjin Hospital, Tianjin, People’s Republic of China
| | - Jinlei Liu
- Cardiology Department, Guang ‘anmen Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, People’s Republic of China
| | - Guangzhong Zhang
- Dermatological Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Xin Feng
- Pharmacy Department, Tianjin Hospital, Tianjin, People’s Republic of China
| | - Wenchao Dan
- Dermatological Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
- Correspondence: Wenchao Dan, Dermatological Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China, Tel +86 13652001152, Email
| |
Collapse
|