1
|
Gonzalez G, Lin X, Herath I, Veselkov K, Bronstein M, Zitnik M. Combinatorial prediction of therapeutic perturbations using causally-inspired neural networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.01.03.573985. [PMID: 38260532 PMCID: PMC10802439 DOI: 10.1101/2024.01.03.573985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Phenotype-driven approaches identify disease-counteracting compounds by analyzing the phenotypic signatures that distinguish diseased from healthy states. These approaches can guide the discovery of targeted perturbations, including small-molecule drugs and genetic interventions, that modulate disease phenotypes toward healthier states. Here, we introduce PDGrapher, a causally inspired graph neural network (GNN) designed to predict combinatorial perturbagens (sets of therapeutic targets) capable of reversing disease phenotypes. Unlike methods that learn how perturbations alter phenotypes, PDGrapher solves the inverse problem of directly predicting the perturbagens needed to achieve a desired response. PDGrapher is a GNN that embeds disease cell states into gene regulatory or protein-protein interaction networks, learns a latent representation of these states, and identifies the optimal combinatorial perturbations that most effectively shift the diseased state toward the desired treated state within that latent space. In experiments in nine cell lines with chemical perturbations, PDGrapher identified effective per-turbagens in up to 13.33% more test samples than competing methods and achieved a normalized discounted cumulative gain of up to 0.12 higher to classify therapeutic targets. It also demonstrated competitive performance on ten genetic perturbation datasets. A key advantage of PDGrapher is its direct prediction paradigm, in contrast to the indirect and computationally intensive models traditionally employed in phenotype-driven research. This approach accelerates training by up to 25 times compared to existing methods. PDGrapher provides a fast approach for identifying therapeutic perturbations and advancing phenotype-driven drug discovery.
Collapse
Affiliation(s)
- Guadalupe Gonzalez
- Imperial College London, London, UK
- Prescient Design, Genentech, South San Francisco, CA, USA
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Xiang Lin
- Harvard Medical School, Boston, MA, USA
| | - Isuru Herath
- Merck & Co., South San Francisco, CA, USA
- Cornell University, Ithaca, NY, USA
| | | | | | - Marinka Zitnik
- Harvard Medical School, Boston, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Data Science Initiative, Cambridge, MA, USA
| |
Collapse
|
2
|
Sun S, Shyr Z, McDaniel K, Fang Y, Tao D, Chen CZ, Zheng W, Zhu Q. Reversal gene expression assessment for drug repurposing, a case study of glioblastoma. J Transl Med 2025; 23:25. [PMID: 39773231 PMCID: PMC11706105 DOI: 10.1186/s12967-024-06046-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a rare brain cancer with an exceptionally high mortality rate, which illustrates the pressing demand for more effective therapeutic options. Despite considerable research efforts on GBM, its underlying biological mechanisms remain unclear. Furthermore, none of the United States Food and Drug Administration (FDA) approved drugs used for GBM deliver satisfactory survival improvement. METHODS This study presents a novel computational pipeline by utilizing gene expression data analysis for GBM for drug repurposing to address the challenges in rare disease drug development, particularly focusing on GBM. The GBM Gene Expression Profile (GGEP) was constructed with multi-omics data to identify drugs with reversal gene expression to GGEP from the Integrated Network-Based Cellular Signatures (iLINCS) database. RESULTS We prioritized the candidates via hierarchical clustering of their expression signatures and quantification of their reversal strength by calculating two self-defined indices based on the GGEP genes' log2 foldchange (LFC) that the drug candidates could induce. Among five prioritized candidates, in-vitro experiments validated Clofarabine and Ciclopirox as highly efficacious in selectively targeting GBM cancer cells. CONCLUSIONS The success of this study illustrated a promising avenue for accelerating drug development by uncovering underlying gene expression effect between drugs and diseases, which can be extended to other rare diseases and non-rare diseases.
Collapse
Affiliation(s)
- Shixue Sun
- Informatics Core, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Zeenat Shyr
- Early Translation Branch, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Kathleen McDaniel
- Early Translation Branch, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Yuhong Fang
- Analytical Chemistry Core, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Dingyin Tao
- Analytical Chemistry Core, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Catherine Z Chen
- Early Translation Branch, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Wei Zheng
- Early Translation Branch, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Qian Zhu
- Informatics Core, Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA.
| |
Collapse
|
3
|
Gao K, Liu L, Lei S, Li Z, Huo P, Wang Z, Dong L, Deng W, Bu D, Zeng X, Li C, Zhao Y, Zhang W, Wang W, Wu Y. HERB 2.0: an updated database integrating clinical and experimental evidence for traditional Chinese medicine. Nucleic Acids Res 2025; 53:D1404-D1414. [PMID: 39558177 PMCID: PMC11701625 DOI: 10.1093/nar/gkae1037] [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: 09/21/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024] Open
Abstract
Clinical trials and meta-analyses are considered high-level medical evidence with solid credibility. However, such clinical evidence for traditional Chinese medicine (TCM) is scattered, requiring a unified entrance to navigate all available evaluations on TCM therapies under modern standards. Besides, novel experimental evidence has continuously accumulated for TCM since the publication of HERB 1.0. Therefore, we updated the HERB database to integrate four types of evidence for TCM: (i) we curated 8558 clinical trials and 8032 meta-analyses information for TCM and extracted clear clinical conclusions for 1941 clinical trials and 593 meta-analyses with companion supporting papers. (ii) we updated experimental evidence for TCM, increased the number of high-throughput experiments to 2231, and curated references to 6 644. We newly added high-throughput experiments for 376 diseases and evaluated all pairwise similarities among TCM herbs/ingredients/formulae, modern drugs and diseases. (iii) we provide an automatic analyzing interface for users to upload their gene expression profiles and map them to our curated datasets. (iv) we built knowledge graph representations of HERB entities and relationships to retrieve TCM knowledge better. In summary, HERB 2.0 represents rich data type, content, utilization, and visualization improvements to support TCM research and guide modern drug discovery. It is accessible through http://herb.ac.cn/v2 or http://47.92.70.12.
Collapse
Affiliation(s)
- Kai Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Liu Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
| | - Shuangshuang Lei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
| | - Zhinong Li
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Peipei Huo
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhihao Wang
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Lei Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
| | - Wenxin Deng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
| | - Dechao Bu
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Chun Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yi Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Wei Zhang
- West China Biomedical Big Data Center, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Wei Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yang Wu
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
4
|
Wei Z, Si D, Duan B, Gao Y, Yu Q, Zhang Z, Guo L, Liu Q. PerturBase: a comprehensive database for single-cell perturbation data analysis and visualization. Nucleic Acids Res 2025; 53:D1099-D1111. [PMID: 39377396 PMCID: PMC11701531 DOI: 10.1093/nar/gkae858] [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: 06/01/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024] Open
Abstract
Single-cell perturbation (scPerturbation) sequencing techniques, represented by single-cell genetic perturbation (e.g. Perturb-seq) and single-cell chemical perturbation (e.g. sci-Plex), result from the integration of single-cell toolkits with conventional bulk screening methods. These innovative sequencing techniques empower researchers to dissect perturbation effects in biological systems at an unprecedented resolution. Despite these advancements, a notable gap exists in the availability of a dedicated database for exploring scPerturbation data. To address this gap, we present PerturBase, the most comprehensive database designed for the analysis and visualization of scPerturbation data (http://www.perturbase.cn/). PerturBase curates 122 datasets from 46 publicly available studies, covering 115 single-modal and 7 multi-modal datasets that include 24 254 genetic and 230 chemical perturbations from approximately 5 million cells. The database, comprising the 'Dataset' and 'Perturbation' modules, provides insights into various results, encompassing quality control, denoising, differential gene expression analysis, functional analysis of perturbation effects and characterization of relationships between perturbations. All the datasets and results are presented on user-friendly, easy-to-browse web pages and can be visualized through intuitive and interactive plot and table formats. In summary, PerturBase stands as a pioneering, high-content database intended for searching, visualizing and analyzing scPerturbation datasets, contributing to a deeper understanding of perturbation effects.
Collapse
Affiliation(s)
- Zhiting Wei
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Duanmiao Si
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Bin Duan
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Yicheng Gao
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Qian Yu
- Zhejiang Lab, Kechuang Avenue, Zhongtai Subdistrict, Yuhang District, Hangzhou 311121, China
| | - Zhenbo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Ling Guo
- Zhejiang Lab, Kechuang Avenue, Zhongtai Subdistrict, Yuhang District, Hangzhou 311121, China
| | - Qi Liu
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
- Zhejiang Lab, Kechuang Avenue, Zhongtai Subdistrict, Yuhang District, Hangzhou 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, 55 Chuanhe Road, Shanghai 200092, China
| |
Collapse
|
5
|
Attri M, Raghav A, Sinha J. Revolutionising Neurological Therapeutics: Investigating Drug Repurposing Strategies. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:115-131. [PMID: 39323347 DOI: 10.2174/0118715273329531240911075309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 09/27/2024]
Abstract
Repurposing drugs (DR) has become a viable approach to hasten the search for cures for neurodegenerative diseases (NDs). This review examines different off-target and on-target drug discovery techniques and how they might be used to find possible treatments for non-diagnostic depressions. Off-target strategies look at the known or unknown side effects of currently approved drugs for repositioning, whereas on-target strategies connect disease pathways to targets that can be treated with drugs. The review highlights the potential of experimental and computational methodologies, such as machine learning, proteomic techniques, network and genomics-based approaches, and in silico screening, in uncovering new drug-disease correlations. It also looks at difficulties and failed attempts at drug repurposing for NDs, highlighting the necessity of exact and standardised procedures to increase success rates. This review's objectives are to address the purpose of drug repurposing in human disorders, particularly neurological diseases, and to provide an overview of repurposing candidates that are presently undergoing clinical trials for neurological conditions, along with any possible causes and early findings. We then include a list of drug repurposing strategies, restrictions, and difficulties for upcoming research.
Collapse
Affiliation(s)
- Meenakshi Attri
- School of Medical & Allied Sciences, K.R. Mangalam University, Gurugram, Haryana 122103, India
| | - Asha Raghav
- Department of Pharmaceutics, School of Health Sciences, Sushant University, Gurugram, Haryana 122003, India
| | - Jyoti Sinha
- Department of Pharmaceutics, School of Health Sciences, Sushant University, Gurugram, Haryana 122003, India
| |
Collapse
|
6
|
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
|
7
|
Yin H, Duo H, Li S, Qin D, Xie L, Xiao Y, Sun J, Tao J, Zhang X, Li Y, Zou Y, Yang Q, Yang X, Hao Y, Li B. Unlocking biological insights from differentially expressed genes: Concepts, methods, and future perspectives. J Adv Res 2024:S2090-1232(24)00560-5. [PMID: 39647635 DOI: 10.1016/j.jare.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/12/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND Identifying differentially expressed genes (DEGs) is a core task of transcriptome analysis, as DEGs can reveal the molecular mechanisms underlying biological processes. However, interpreting the biological significance of large DEG lists is challenging. Currently, gene ontology, pathway enrichment and protein-protein interaction analysis are common strategies employed by biologists. Additionally, emerging analytical strategies/approaches (such as network module analysis, knowledge graph, drug repurposing, cell marker discovery, trajectory analysis, and cell communication analysis) have been proposed. Despite these advances, comprehensive guidelines for systematically and thoroughly mining the biological information within DEGs remain lacking. AIM OF REVIEW This review aims to provide an overview of essential concepts and methodologies for the biological interpretation of DEGs, enhancing the contextual understanding. It also addresses the current limitations and future perspectives of these approaches, highlighting their broad applications in deciphering the molecular mechanism of complex diseases and phenotypes. To assist users in extracting insights from extensive datasets, especially various DEG lists, we developed DEGMiner (https://www.ciblab.net/DEGMiner/), which integrates over 300 easily accessible databases and tools. KEY SCIENTIFIC CONCEPTS OF REVIEW This review offers strong support and guidance for exploring DEGs, and also will accelerate the discovery of hidden biological insights within genomes.
Collapse
Affiliation(s)
- Huachun Yin
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China; Department of Neurosurgery, Xinqiao Hospital, The Army Medical University, Chongqing 400037, PR China; Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, The Army Medical University, Chongqing 400038, PR China
| | - Hongrui Duo
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Song Li
- Department of Neurosurgery, Xinqiao Hospital, The Army Medical University, Chongqing 400037, PR China
| | - Dan Qin
- Department of Biology, College of Science, Northeastern University, Boston, MA 02115, USA
| | - Lingling Xie
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Yingxue Xiao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Jing Sun
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Jingxin Tao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Xiaoxi Zhang
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Yinghong Li
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, PR China
| | - Yue Zou
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Qingxia Yang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Xian Yang
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Youjin Hao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China.
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China.
| |
Collapse
|
8
|
Chowdary R, Suter RK, D'Antuono M, Gomes C, Stein J, Lee KB, Lee JK, Ayad NG. OrthologAL: A Shiny application for quality-aware humanization of non-human pre-clinical high-dimensional gene expression data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625000. [PMID: 39651293 PMCID: PMC11623543 DOI: 10.1101/2024.11.25.625000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Single-cell and spatial transcriptomics provide unprecedented insight into the inner workings of disease. Pharmacotranscriptomic approaches are powerful tools that leverage gene expression data for drug repurposing and treatment discovery in many diseases. Multiple databases attempt to connect human cellular transcriptional responses to small molecules for use in transcriptome-based drug discovery efforts. However, pre-clinical research often requires in vivo experiments in non-human species, which makes capitalizing on such valuable resources difficult. To facilitate the application of pharmacotranscriptomic databases to pre-clinical research models and to facilitate human orthologous conversion of non-human transcriptomes, we introduce OrthologAL. OrthologAL leverages the BioMart database to access different gene sets from Ensembl, facilitating the interaction between these servers without needing user-generated code. Researchers can input their single-cell or other high-dimensional gene expression data from any species, and OrthologAL will output a human ortholog-converted dataset for download and use. To demonstrate the utility of this application, we characterized orthologous conversion in single-cell, single-nuclei, and spatial transcriptomic data derived from common pre-clinical models, including patient-derived orthotopic xenografts of medulloblastoma, and mouse and rat models of spinal cord injury. We show that OrthologAL can convert these data types efficiently to that of corresponding orthologs while preserving the dimensional architecture of the original non-human expression data. OrthologAL will be broadly useful for applying pre-clinical, high-dimensional transcriptomics data in functional small molecule predictions using existing human-annotated databases.
Collapse
|
9
|
Liao J, Yi H, Wang H, Yang S, Jiang D, Huang X, Zhang M, Shen J, Lu H, Niu Y. CDCM: a correlation-dependent connectivity map approach to rapidly screen drugs during outbreaks of infectious diseases. Brief Bioinform 2024; 26:bbae659. [PMID: 39701599 DOI: 10.1093/bib/bbae659] [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: 07/02/2024] [Revised: 09/06/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
In the context of the global damage caused by coronavirus disease 2019 (COVID-19) and the emergence of the monkeypox virus (MPXV) outbreak as a public health emergency of international concern, research into methods that can rapidly test potential therapeutics during an outbreak of a new infectious disease is urgently needed. Computational drug discovery is an effective way to solve such problems. The existence of various large open databases has mitigated the time and resource consumption of traditional drug development and improved the speed of drug discovery. However, the diversity of cell lines used in various databases remains limited, and previous drug discovery methods are ineffective for cross-cell prediction. In this study, we propose a correlation-dependent connectivity map (CDCM) to achieve cross-cell predictions of drug similarity. The CDCM mainly identifies drug-drug or disease-drug relationships from the perspective of gene networks by exploring the correlation changes between genes and identifying similarities in the effects of drugs or diseases on gene expression. We validated the CDCM on multiple datasets and found that it performed well for drug identification across cell lines. A comparison with the Connectivity Map revealed that our method was more stable and performed better across different cell lines. In the application of the CDCM to COVID-19 and MPXV data, the predictions of potential therapeutic compounds for COVID-19 were consistent with several previous studies, and most of the predicted drugs were found to be experimentally effective against MPXV. This result confirms the practical value of the CDCM. With the ability to predict across cell lines, the CDCM outperforms the Connectivity Map, and it has wider application prospects and a reduced cost of use.
Collapse
Affiliation(s)
- Junlei Liao
- School of Mathematics and Statistics, HNP-LAMA, Central South University, Changsha 410083, Hunan, China
| | - Hongyang Yi
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China
| | - Hao Wang
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen 518133, China
| | - Sumei Yang
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China
| | - Duanmei Jiang
- School of Mathematics and Statistics, HNP-LAMA, Central South University, Changsha 410083, Hunan, China
| | - Xin Huang
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen 518133, China
| | - Mingxia Zhang
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China
| | - Jiayin Shen
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China
| | - Hongzhou Lu
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China
| | - Yuanling Niu
- School of Mathematics and Statistics, HNP-LAMA, Central South University, Changsha 410083, Hunan, China
| |
Collapse
|
10
|
Guo K, Yang J, Jiang R, Ren X, Liu P, Wang W, Zhou S, Wang X, Ma L, Hu Y. Identification of Key Immune and Cell Cycle Modules and Prognostic Genes for Glioma Patients through Transcriptome Analysis. Pharmaceuticals (Basel) 2024; 17:1295. [PMID: 39458936 PMCID: PMC11514598 DOI: 10.3390/ph17101295] [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: 07/16/2024] [Revised: 09/06/2024] [Accepted: 09/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Gliomas, the most prevalent type of primary brain tumor, stand out as one of the most aggressive and lethal types of human cancer. METHODS & RESULTS To uncover potential prognostic markers, we employed the weighted correlation network analysis (WGCNA) on the Chinese Glioma Genome Atlas (CGGA) 693 dataset to reveal four modules significantly associated with glioma clinical traits, primarily involved in immune function, cell cycle regulation, and ribosome biogenesis. Using the least absolute shrinkage and selection operator (LASSO) regression algorithm, we identified 11 key genes and developed a prognostic risk score model, which exhibits precise prognostic prediction in the CGGA 325 dataset. More importantly, we also validated the model in 12 glioma patients with overall survival (OS) ranging from 4 to 132 months using mRNA sequencing and immunohistochemical analysis. The analysis of immune infiltration revealed that patients with high-risk scores exhibit a heightened immune infiltration, particularly immune suppression cells, along with increased expression of immune checkpoints. Furthermore, we explored potentially effective drugs targeting 11 key genes for gliomas using the library of integrated network-based cellular signatures (LINCS) L1000 database, identifying that in vitro, both torin-1 and clofarabine exhibit promising anti-glioma activity and inhibitory effect on the cell cycle, a significant pathway enriched in the identified glioma modules. CONCLUSIONS In conclusion, our study provides valuable insights into molecular mechanisms and identifying potential therapeutic targets for gliomas.
Collapse
Affiliation(s)
- Kaimin Guo
- Tianjin Tasly Digital Intelligence Chinese Medicine Development Co., Ltd., Tianjin 300410, China; (K.G.); (J.Y.); (X.R.); (W.W.); (S.Z.)
- State Key Laboratory of Chinese Medicine Modernization, Tianjin 300193, China;
| | - Jinna Yang
- Tianjin Tasly Digital Intelligence Chinese Medicine Development Co., Ltd., Tianjin 300410, China; (K.G.); (J.Y.); (X.R.); (W.W.); (S.Z.)
- State Key Laboratory of Chinese Medicine Modernization, Tianjin 300193, China;
| | - Ruonan Jiang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China;
| | - Xiaxia Ren
- Tianjin Tasly Digital Intelligence Chinese Medicine Development Co., Ltd., Tianjin 300410, China; (K.G.); (J.Y.); (X.R.); (W.W.); (S.Z.)
- State Key Laboratory of Chinese Medicine Modernization, Tianjin 300193, China;
| | - Peng Liu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin 300193, China;
| | - Wenjia Wang
- Tianjin Tasly Digital Intelligence Chinese Medicine Development Co., Ltd., Tianjin 300410, China; (K.G.); (J.Y.); (X.R.); (W.W.); (S.Z.)
- State Key Laboratory of Chinese Medicine Modernization, Tianjin 300193, China;
| | - Shuiping Zhou
- Tianjin Tasly Digital Intelligence Chinese Medicine Development Co., Ltd., Tianjin 300410, China; (K.G.); (J.Y.); (X.R.); (W.W.); (S.Z.)
- State Key Laboratory of Chinese Medicine Modernization, Tianjin 300193, China;
| | - Xiaoguang Wang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China;
| | - Li Ma
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China;
| | - Yunhui Hu
- Tianjin Tasly Digital Intelligence Chinese Medicine Development Co., Ltd., Tianjin 300410, China; (K.G.); (J.Y.); (X.R.); (W.W.); (S.Z.)
- State Key Laboratory of Chinese Medicine Modernization, Tianjin 300193, China;
| |
Collapse
|
11
|
Sun S, Shyr Z, McDaniel K, Fang Y, Tao D, Chen CZ, Zheng W, Zhu Q. Reversal Gene Expression Assessment for Drug Repurposing, a Case Study of Glioblastoma. RESEARCH SQUARE 2024:rs.3.rs-4765282. [PMID: 39315277 PMCID: PMC11419258 DOI: 10.21203/rs.3.rs-4765282/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Glioblastoma (GBM) is a rare brain cancer with an exceptionally high mortality rate, which illustrates the pressing demand for more effective therapeutic options. Despite considerable research efforts on GBM, its underlying biological mechanisms remain unclear. Furthermore, none of the United States Food and Drug Administration (FDA) approved drugs used for GBM deliver satisfactory survival improvement. This study presents a novel computational pipeline by utilizing gene expression data analysis for GBM for drug repurposing to address the challenges in rare disease drug development, particularly focusing on GBM. The GBM Gene Expression Profile (GGEP) was constructed with multi-omics data to identify drugs with reversal gene expression to GGEP from the Integrated Network-Based Cellular Signatures (iLINCS) database. We prioritized the candidates via hierarchical clustering of their expression signatures and quantification of their reversal strength by calculating two self-defined indices based on the GGEP genes' log2 foldchange (LFCs) that the drug candidates could induce. Among eight prioritized candidates, in-vitro experiments validated Clofarabine and Ciclopirox as highly efficacious in selectively targeting GBM cancer cells. The success of this study illustrated a promising avenue for accelerating drug development by uncovering underlying gene expression effect between drugs and diseases, which can be extended to other rare diseases and non-rare diseases.
Collapse
Affiliation(s)
- Shixue Sun
- NCATS: National Center for Advancing Translational Sciences
| | - Zeenat Shyr
- NCATS: National Center for Advancing Translational Sciences
| | - Kathleen McDaniel
- NCATS ETB: National Center for Advancing Translational Sciences Early Translation Branch
| | - Yuhong Fang
- NCATS: National Center for Advancing Translational Sciences
| | - Dingyin Tao
- NCATS: National Center for Advancing Translational Sciences
| | | | - Wei Zheng
- NCATS: National Center for Advancing Translational Sciences
| | - Qian Zhu
- NCATS: National Center for Advancing Translational Sciences
| |
Collapse
|
12
|
Kenakin T. Know your molecule: pharmacological characterization of drug candidates to enhance efficacy and reduce late-stage attrition. Nat Rev Drug Discov 2024; 23:626-644. [PMID: 38890494 DOI: 10.1038/s41573-024-00958-9] [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: 04/23/2024] [Indexed: 06/20/2024]
Abstract
Despite advances in chemical, computational and biological sciences, the rate of attrition of drug candidates in clinical development is still high. A key point in the small-molecule discovery process that could provide opportunities to help address this challenge is the pharmacological characterization of hit and lead compounds, culminating in the selection of a drug candidate. Deeper characterization is increasingly important, because the 'quality' of drug efficacy, at least for G protein-coupled receptors (GPCRs), is now understood to be much more than activation of commonly evaluated pathways such as cAMP signalling, with many more 'efficacies' of ligands that could be harnessed therapeutically. Such characterization is being enabled by novel assays to characterize the complex behaviour of GPCRs, such as biased signalling and allosteric modulation, as well as advances in structural biology, such as cryo-electron microscopy. This article discusses key factors in the assessments of the pharmacology of hit and lead compounds in the context of GPCRs as a target class, highlighting opportunities to identify drug candidates with the potential to address limitations of current therapies and to improve the probability of them succeeding in clinical development.
Collapse
Affiliation(s)
- Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
13
|
Zhang W, Huang RS. Computer-aided drug discovery strategies for novel therapeutics for prostate cancer leveraging next-generating sequencing data. Expert Opin Drug Discov 2024; 19:841-853. [PMID: 38860709 PMCID: PMC11537242 DOI: 10.1080/17460441.2024.2365370] [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: 03/14/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
INTRODUCTION Prostate cancer (PC) is the most common malignancy and accounts for a significant proportion of cancer deaths among men. Although initial therapy success can often be observed in patients diagnosed with localized PC, many patients eventually develop disease recurrence and metastasis. Without effective treatments, patients with aggressive PC display very poor survival. To curb the current high mortality rate, many investigations have been carried out to identify efficacious therapeutics. Compared to de novo drug designs, computational methods have been widely employed to offer actionable drug predictions in a fast and cost-efficient way. Particularly, powered by an increasing availability of next-generation sequencing molecular profiles from PC patients, computer-aided approaches can be tailored to screen for candidate drugs. AREAS COVERED Herein, the authors review the recent advances in computational methods for drug discovery utilizing molecular profiles from PC patients. Given the uniqueness in PC therapeutic needs, they discuss in detail the drug discovery goals of these studies, highlighting their translational values for clinically impactful drug nomination. EXPERT OPINION Evolving molecular profiling techniques may enable new perspectives for computer-aided approaches to offer drug candidates for different tumor microenvironments. With ongoing efforts to incorporate new compounds into large-scale high-throughput screens, the authors envision continued expansion of drug candidate pools.
Collapse
Affiliation(s)
- Weijie Zhang
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - R. Stephanie Huang
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
14
|
Tong X, Qu N, Kong X, Ni S, Zhou J, Wang K, Zhang L, Wen Y, Shi J, Zhang S, Li X, Zheng M. Deep representation learning of chemical-induced transcriptional profile for phenotype-based drug discovery. Nat Commun 2024; 15:5378. [PMID: 38918369 PMCID: PMC11199551 DOI: 10.1038/s41467-024-49620-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Artificial intelligence transforms drug discovery, with phenotype-based approaches emerging as a promising alternative to target-based methods, overcoming limitations like lack of well-defined targets. While chemical-induced transcriptional profiles offer a comprehensive view of drug mechanisms, inherent noise often obscures the true signal, hindering their potential for meaningful insights. Here, we highlight the development of TranSiGen, a deep generative model employing self-supervised representation learning. TranSiGen analyzes basal cell gene expression and molecular structures to reconstruct chemical-induced transcriptional profiles with high accuracy. By capturing both cellular and compound information, TranSiGen-derived representations demonstrate efficacy in diverse downstream tasks like ligand-based virtual screening, drug response prediction, and phenotype-based drug repurposing. Notably, in vitro validation of TranSiGen's application in pancreatic cancer drug discovery highlights its potential for identifying effective compounds. We envisage that integrating TranSiGen into the drug discovery and mechanism research holds significant promise for advancing biomedicine.
Collapse
Affiliation(s)
- Xiaochu Tong
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Ning Qu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Xiangtai Kong
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Shengkun Ni
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Jingyi Zhou
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Lingang Laboratory, Shanghai, 200031, China
| | - Kun Wang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Lehan Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Yiming Wen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Jiangshan Shi
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Sulin Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
| | - Xutong Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
15
|
Lee DH, Yoo JK, Um KH, Ha W, Lee SM, Park J, Kye MJ, Suh J, Choi JW. Intravesical instillation-based mTOR-STAT3 dual targeting for bladder cancer treatment. J Exp Clin Cancer Res 2024; 43:170. [PMID: 38886756 PMCID: PMC11184849 DOI: 10.1186/s13046-024-03088-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Recent intravesical administration of adenoviral vectors, either as a single injection or in combination with immune checkpoint inhibitors, exemplified by cretostimogene grenadenorepvec and nadofaragene firadenovec, has demonstrated remarkable efficacy in clinical trials for non-muscle invasive bladder cancer. Despite their ability to induce an enhanced immune reaction within the lesion, the intracellular survival signaling of cancer cells has not been thoroughly addressed. METHODS An analysis of the prognostic data revealed a high probability of therapeutic efficacy with simultaneous inhibition of mTOR and STAT3. Considering the challenges of limited pharmaco-accessibility to the bladder due to its pathophysiological structure and the partially undruggable nature of target molecules, we designed a dual siRNA system targeting both mRNAs. Subsequently, this dual siRNA system was encoded into the adenovirus 5/3 (Ad 5/3) to enhance in vivo delivery efficiency. RESULTS Gene-targeting efficacy was assessed using cells isolated from xenografted tumors using a single-cell analysis system. Our strategy demonstrated a balanced downregulation of mTOR and STAT3 at the single-cell resolution, both in vitro and in vivo. This approach reduced tumor growth in bladder cancer xenograft and orthotopic animal experiments. In addition, increased infiltration of CD8+ T cells was observed in a humanized mouse model. We provided helpful and safe tissue distribution data for intravesical therapy of siRNAs coding adenoviruses. CONCLUSIONS The bi-specific siRNA strategy, encapsulated in an adenovirus, could be a promising tool to augment cancer treatment efficacy and overcome conventional therapy limitations associated with "undruggability." Hence, we propose that dual targeting of mTOR and STAT3 is an advantageous strategy for intravesical therapy using adenoviruses.
Collapse
Affiliation(s)
- Dae Hoon Lee
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- R&D Center of Curigin Ltd., Curigin, Seoul, 04778, Republic of Korea
| | - Jung Ki Yoo
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- R&D Center of Curigin Ltd., Curigin, Seoul, 04778, Republic of Korea
| | - Ki Hwan Um
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- R&D Center of Curigin Ltd., Curigin, Seoul, 04778, Republic of Korea
- Department of Regulatory Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Wootae Ha
- R&D Center of Curigin Ltd., Curigin, Seoul, 04778, Republic of Korea
| | - Soo Min Lee
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Regulatory Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Junseong Park
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Min Jeong Kye
- R&D Center of Curigin Ltd., Curigin, Seoul, 04778, Republic of Korea
| | - Jungyo Suh
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Jin Woo Choi
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
- R&D Center of Curigin Ltd., Curigin, Seoul, 04778, Republic of Korea.
- Department of Regulatory Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
16
|
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
|
17
|
Fisher JL, Wilk EJ, Oza VH, Gary SE, Howton TC, Flanary VL, Clark AD, Hjelmeland AB, Lasseigne BN. Signature reversion of three disease-associated gene signatures prioritizes cancer drug repurposing candidates. FEBS Open Bio 2024; 14:803-830. [PMID: 38531616 PMCID: PMC11073506 DOI: 10.1002/2211-5463.13796] [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: 02/11/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Drug repurposing is promising because approving a drug for a new indication requires fewer resources than approving a new drug. Signature reversion detects drug perturbations most inversely related to the disease-associated gene signature to identify drugs that may reverse that signature. We assessed the performance and biological relevance of three approaches for constructing disease-associated gene signatures (i.e., limma, DESeq2, and MultiPLIER) and prioritized the resulting drug repurposing candidates for four low-survival human cancers. Our results were enriched for candidates that had been used in clinical trials or performed well in the PRISM drug screen. Additionally, we found that pamidronate and nimodipine, drugs predicted to be efficacious against the brain tumor glioblastoma (GBM), inhibited the growth of a GBM cell line and cells isolated from a patient-derived xenograft (PDX). Our results demonstrate that by applying multiple disease-associated gene signature methods, we prioritized several drug repurposing candidates for low-survival cancers.
Collapse
Affiliation(s)
- Jennifer L. Fisher
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Elizabeth J. Wilk
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Vishal H. Oza
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Sam E. Gary
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Timothy C. Howton
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Victoria L. Flanary
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Amanda D. Clark
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Anita B. Hjelmeland
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| |
Collapse
|
18
|
Rivas SR, Mendez Valdez MJ, Chandar JS, Desgraves JF, Lu VM, Ampie L, Singh EB, Seetharam D, Ramsoomair CK, Hudson A, Ingle SM, Govindarajan V, Doucet-O’Hare TT, DeMarino C, Heiss JD, Nath A, Shah AH. Antiretroviral Drug Repositioning for Glioblastoma. Cancers (Basel) 2024; 16:1754. [PMID: 38730705 PMCID: PMC11083594 DOI: 10.3390/cancers16091754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/13/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Outcomes for glioblastoma (GBM) remain poor despite standard-of-care treatments including surgical resection, radiation, and chemotherapy. Intratumoral heterogeneity contributes to treatment resistance and poor prognosis, thus demanding novel therapeutic approaches. Drug repositioning studies on antiretroviral therapy (ART) have shown promising potent antineoplastic effects in multiple cancers; however, its efficacy in GBM remains unclear. To better understand the pleiotropic anticancer effects of ART on GBM, we conducted a comprehensive drug repurposing analysis of ART in GBM to highlight its utility in translational neuro-oncology. To uncover the anticancer role of ART in GBM, we conducted a comprehensive bioinformatic and in vitro screen of antiretrovirals against glioblastoma. Using the DepMap repository and reversal of gene expression score, we conducted an unbiased screen of 16 antiretrovirals in 40 glioma cell lines to identify promising candidates for GBM drug repositioning. We utilized patient-derived neurospheres and glioma cell lines to assess neurosphere viability, proliferation, and stemness. Our in silico screen revealed that several ART drugs including reverse transcriptase inhibitors (RTIs) and protease inhibitors (PIs) demonstrated marked anti-glioma activity with the capability of reversing the GBM disease signature. RTIs effectively decreased cell viability, GBM stem cell markers, and proliferation. Our study provides mechanistic and functional insight into the utility of ART repurposing for malignant gliomas, which supports the current literature. Given their safety profile, preclinical efficacy, and neuropenetrance, ARTs may be a promising adjuvant treatment for GBM.
Collapse
Affiliation(s)
- Sarah R. Rivas
- Surgical Neurology Branch, National Institute of Neurological Diseases and Stroke, Bethesda, MD 20892, USA; (S.R.R.); (L.A.); (A.N.)
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| | - Mynor J. Mendez Valdez
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| | - Jay S. Chandar
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| | - Jelisah F. Desgraves
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| | - Victor M. Lu
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| | - Leo Ampie
- Surgical Neurology Branch, National Institute of Neurological Diseases and Stroke, Bethesda, MD 20892, USA; (S.R.R.); (L.A.); (A.N.)
| | - Eric B. Singh
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| | - Deepa Seetharam
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| | - Christian K. Ramsoomair
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| | - Anna Hudson
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| | - Shreya M. Ingle
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| | - Vaidya Govindarajan
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| | - Tara T. Doucet-O’Hare
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Catherine DeMarino
- Surgical Neurology Branch, National Institute of Neurological Diseases and Stroke, Bethesda, MD 20892, USA; (S.R.R.); (L.A.); (A.N.)
| | - John D. Heiss
- Surgical Neurology Branch, National Institute of Neurological Diseases and Stroke, Bethesda, MD 20892, USA; (S.R.R.); (L.A.); (A.N.)
| | - Avindra Nath
- Surgical Neurology Branch, National Institute of Neurological Diseases and Stroke, Bethesda, MD 20892, USA; (S.R.R.); (L.A.); (A.N.)
| | - Ashish H. Shah
- Section of Virology and Immunotherapy, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA (E.B.S.)
| |
Collapse
|
19
|
Qian ZY, Pan YQ, Li XX, Chen YX, Wu HX, Liu ZX, Kosar M, Bartek J, Wang ZX, Xu RH. Modulator of TMB-associated immune infiltration (MOTIF) predicts immunotherapy response and guides combination therapy. Sci Bull (Beijing) 2024; 69:803-822. [PMID: 38320897 DOI: 10.1016/j.scib.2024.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/04/2023] [Accepted: 12/07/2023] [Indexed: 02/08/2024]
Abstract
Patients with high tumor mutational burden (TMB) levels do not consistently respond to immune checkpoint inhibitors (ICIs), possibly because a high TMB level does not necessarily result in adequate infiltration of CD8+ T cells. Using bulk ribonucleic acid sequencing (RNA-seq) data from 9311 tumor samples across 30 cancer types, we developed a novel tool called the modulator of TMB-associated immune infiltration (MOTIF), which comprises genes that can determine the extent of CD8+ T cell infiltration prompted by a certain TMB level. We confirmed that MOTIF can accurately reflect the integrity and defects of the cancer-immunity cycle. By analyzing 84 human single-cell RNA-seq datasets from 32 types of solid tumors, we revealed that MOTIF can provide insights into the diverse roles of various cell types in the modulation of CD8+ T cell infiltration. Using pretreatment RNA-seq data from 13 ICI-treated cohorts, we validated the use of MOTIF in predicting CD8+ T cell infiltration and ICI efficacy. Among the components of MOTIF, we identified EMC3 as a negative regulator of CD8+ T cell infiltration, which was validated via in vivo studies. Additionally, MOTIF provided guidance for the potential combinations of programmed death 1 blockade with certain immunostimulatory drugs to facilitate CD8+ T cell infiltration and improve ICI efficacy.
Collapse
Affiliation(s)
- Zheng-Yu Qian
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Yi-Qian Pan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Xue-Xin Li
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm S-171 21, Sweden; Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Yan-Xing Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Hao-Xiang Wu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Ze-Xian Liu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China; Bioinformatics Platform, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Laboratory of Artificial Intelligence and Data Science, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Martin Kosar
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm S-171 21, Sweden; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining 314400, China; Edinburgh Medical School, Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH1 1LT, UK
| | - Jiri Bartek
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm S-171 21, Sweden; Danish Cancer Society Research Center, Copenhagen DK-2100, Denmark.
| | - Zi-Xian Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China; Laboratory of Artificial Intelligence and Data Science, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China; Laboratory of Artificial Intelligence and Data Science, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
20
|
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
|
21
|
Atri P, Shah A, Natarajan G, Rachagani S, Rauth S, Ganguly K, Carmicheal J, Ghersi D, Cox JL, Smith LM, Jain M, Kumar S, Ponnusamy MP, Seshacharyulu P, Batra SK. Connectivity mapping-based identification of pharmacological inhibitor targeting HDAC6 in aggressive pancreatic ductal adenocarcinoma. NPJ Precis Oncol 2024; 8:66. [PMID: 38454151 PMCID: PMC10920818 DOI: 10.1038/s41698-024-00562-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains highly lethal due to limited therapeutic options and expensive/burdensome drug discovery processes. Utilizing genomic-data-driven Connectivity Mapping (CMAP) to identify a drug closer to real-world PC targeting may improve pancreatic cancer (PC) patient outcomes. Initially, we mapped CMAP data to gene expression from 106 PC patients, identifying nine negatively connected drugs. These drugs were further narrowed down using a similar analysis for PC cell lines, human tumoroids, and patient-derived xenografts datasets, where ISOX emerged as the most potent agent to target PC. We used human and mouse syngeneic PC cells, human and mouse tumoroids, and in vivo mice to assess the ability of ISOX alone and in combination with 5FU to inhibit tumor growth. Global transcriptomic and pathway analysis of the ISOX-LINCS signature identified HDAC 6/cMyc as the target axis for ISOX. Specifically, we discovered that genetic and pharmacological targeting of HDAC 6 affected non-histone protein cMyc acetylation, leading to cMyc instability, thereby disrupting PC growth and metastasis by affecting cancer stemness. Finally, KrasG12D harboring tumoroids and mice responded effectively against ISOX and 5FU treatment by enhancing survival and controlling metastasis incidence. Overall, our data validate ISOX as a new drug to treat advanced PC patients without toxicity to normal cells. Our study supports the clinical utility of ISOX along with 5FU in future PC clinical trials.
Collapse
Affiliation(s)
- Pranita Atri
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gopalakrishnan Natarajan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Koelina Ganguly
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Joseph Carmicheal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dario Ghersi
- School of Interdisciplinary Informatics, College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE, USA
| | - Jesse L Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lynette M Smith
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
22
|
Pang Z, Cravatt BF, Ye L. Deciphering Drug Targets and Actions with Single-Cell and Spatial Resolution. Annu Rev Pharmacol Toxicol 2024; 64:507-526. [PMID: 37722721 DOI: 10.1146/annurev-pharmtox-033123-123610] [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: 09/20/2023]
Abstract
Recent advances in chemical, molecular, and genetic approaches have provided us with an unprecedented capacity to identify drug-target interactions across the whole proteome and genome. Meanwhile, rapid developments of single-cell and spatial omics technologies are revolutionizing our understanding of the molecular architecture of biological systems. However, a significant gap remains in how we align our understanding of drug actions, traditionally based on molecular affinities, with the in vivo cellular and spatial tissue heterogeneity revealed by these newer techniques. Here, we review state-of-the-art methods for profiling drug-target interactions and emerging multiomics tools to delineate the tissue heterogeneity at single-cell resolution. Highlighting the recent technical advances enabling high-resolution, multiplexable in situ small-molecule drug imaging (clearing-assisted tissue click chemistry, or CATCH), we foresee the integration of single-cell and spatial omics platforms, data, and concepts into the future framework of defining and understanding in vivo drug-target interactions and mechanisms of actions.
Collapse
Affiliation(s)
- Zhengyuan Pang
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA;
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, California, USA;
| | - Li Ye
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA;
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
23
|
Hou D, Lin H, Feng Y, Zhou K, Li X, Yang Y, Wang S, Yang X, Wang J, Zhao H, Zhang X, Fan J, Lu S, Wang D, Zhu L, Ju D, Chen YZ, Zeng X. CMAUP database update 2024: extended functional and association information of useful plants for biomedical research. Nucleic Acids Res 2024; 52:D1508-D1518. [PMID: 37897343 PMCID: PMC10767869 DOI: 10.1093/nar/gkad921] [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: 09/05/2023] [Revised: 09/23/2023] [Accepted: 10/10/2023] [Indexed: 10/30/2023] Open
Abstract
Knowledge of the collective activities of individual plants together with the derived clinical effects and targeted disease associations is useful for plant-based biomedical research. To provide the information in complement to the established databases, we introduced a major update of CMAUP database, previously featured in NAR. This update includes (i) human transcriptomic changes overlapping with 1152 targets of 5765 individual plants, covering 74 diseases from 20 027 patient samples; (ii) clinical information for 185 individual plants in 691 clinical trials; (iii) drug development information for 4694 drug-producing plants with metabolites developed into approved or clinical trial drugs; (iv) plant and human disease associations (428 737 associations by target, 220 935 reversion of transcriptomic changes, 764 and 154121 associations by clinical trials of individual plants and plant ingredients); (v) the location of individual plants in the phylogenetic tree for navigating taxonomic neighbors, (vi) DNA barcodes of 3949 plants, (vii) predicted human oral bioavailability of plant ingredients by the established SwissADME and HobPre algorithm, (viii) 21-107% increase of CMAUP data over the previous version to cover 60 222 chemical ingredients, 7865 plants, 758 targets, 1399 diseases, 238 KEGG human pathways, 3013 gene ontologies and 1203 disease ontologies. CMAUP update version is freely accessible at https://bidd.group/CMAUP/index.html.
Collapse
Affiliation(s)
- Dongyue Hou
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Hanbo Lin
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Yuhan Feng
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Kaicheng Zhou
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Xingxiu Li
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Yuan Yang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Shuaiqi Wang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Xue Yang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Jiayu Wang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Hui Zhao
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Xuyao Zhang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Jiajun Fan
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - SongLin Lu
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Dan Wang
- Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Lyuhan Zhu
- Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| | - Yu Zong Chen
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Xian Zeng
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai 201203, China
| |
Collapse
|
24
|
Bhowmick C, Rahaman M, Bhattacharya S, Mukherjee M, Chakravorty N, Dutta PK, Mahadevappa M. Identification of hub genes to determine drug-disease correlation in breast carcinomas. Med Oncol 2023; 41:36. [PMID: 38153604 DOI: 10.1007/s12032-023-02246-9] [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: 08/26/2023] [Accepted: 11/11/2023] [Indexed: 12/29/2023]
Abstract
The exact molecular mechanism underlying the heterogeneous drug response against breast carcinoma remains to be fully understood. It is urgently required to identify key genes that are intricately associated with varied clinical response of standard anti-cancer drugs, clinically used to treat breast cancer patients. In the present study, the utility of transcriptomic data of breast cancer patients in discerning the clinical drug response using machine learning-based approaches were evaluated. Here, a computational framework has been developed which can be used to identify key genes that can be linked with clinical drug response and progression of cancer, offering an immense opportunity to predict potential prognostic biomarkers and therapeutic targets. The framework concerned utilizes DeSeq2, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Cytoscape, and machine learning techniques to find these crucial genes. Total RNA extraction and qRT-PCR were performed to quantify relative expression of few hub genes selected from the networks. In our study, we have experimentally checked the expression of few key hub genes like APOA2, DLX5, APOC3, CAMK2B, and PAK6 that were predicted to play an immense role in breast cancer tumorigenesis and progression in response to anti-cancer drug Paclitaxel. However, further experimental validations will be required to get mechanistic insights of these genes in regulating the drug response and cancer progression which will likely to play pivotal role in cancer treatment and precision oncology.
Collapse
Affiliation(s)
- Chiranjib Bhowmick
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Medinipur, Kharagpur, West Bengal, 721302, India
| | - Motiur Rahaman
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Medinipur, Kharagpur, West Bengal, 721302, India
| | - Shatarupa Bhattacharya
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Medinipur, Kharagpur, West Bengal, 721302, India
| | - Mandrita Mukherjee
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Medinipur, Kharagpur, West Bengal, 721302, India
| | - Nishant Chakravorty
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Medinipur, Kharagpur, West Bengal, 721302, India
| | - Pranab Kumar Dutta
- Department of Electrical Engineering, Indian Institute of Technology Kharagpur, West Medinipur, Kharagpur, West Bengal, 721302, India
| | - Manjunatha Mahadevappa
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Medinipur, Kharagpur, West Bengal, 721302, India.
| |
Collapse
|
25
|
Caputo WL, de Souza MC, Basso CR, Pedrosa VDA, Seiva FRF. Comprehensive Profiling and Therapeutic Insights into Differentially Expressed Genes in Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:5653. [PMID: 38067357 PMCID: PMC10705715 DOI: 10.3390/cancers15235653] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 02/16/2024] Open
Abstract
Background: Drug repurposing is a strategy that complements the conventional approach of developing new drugs. Hepatocellular carcinoma (HCC) is a highly prevalent type of liver cancer, necessitating an in-depth understanding of the underlying molecular alterations for improved treatment. Methods: We searched for a vast array of microarray experiments in addition to RNA-seq data. Through rigorous filtering processes, we have identified highly representative differentially expressed genes (DEGs) between tumor and non-tumor liver tissues and identified a distinct class of possible new candidate drugs. Results: Functional enrichment analysis revealed distinct biological processes associated with metal ions, including zinc, cadmium, and copper, potentially implicating chronic metal ion exposure in tumorigenesis. Conversely, up-regulated genes are associated with mitotic events and kinase activities, aligning with the relevance of kinases in HCC. To unravel the regulatory networks governing these DEGs, we employed topological analysis methods, identifying 25 hub genes and their regulatory transcription factors. In the pursuit of potential therapeutic options, we explored drug repurposing strategies based on computational approaches, analyzing their potential to reverse the expression patterns of key genes, including AURKA, CCNB1, CDK1, RRM2, and TOP2A. Potential therapeutic chemicals are alvocidib, AT-7519, kenpaullone, PHA-793887, JNJ-7706621, danusertibe, doxorubicin and analogues, mitoxantrone, podofilox, teniposide, and amonafide. Conclusion: This multi-omic study offers a comprehensive view of DEGs in HCC, shedding light on potential therapeutic targets and drug repurposing opportunities.
Collapse
Affiliation(s)
- Wesley Ladeira Caputo
- Post Graduation Program in Experimental Pathology, State University of Londrina (UEL), Londrina 86057-970, PR, Brazil; (W.L.C.); (M.C.d.S.)
| | - Milena Cremer de Souza
- Post Graduation Program in Experimental Pathology, State University of Londrina (UEL), Londrina 86057-970, PR, Brazil; (W.L.C.); (M.C.d.S.)
| | - Caroline Rodrigues Basso
- Department of Chemical and Biological Sciences, Institute of Bioscience, São Paulo State University (UNESP), Botucatu 18610-034, SP, Brazil; (C.R.B.); (V.d.A.P.)
| | - Valber de Albuquerque Pedrosa
- Department of Chemical and Biological Sciences, Institute of Bioscience, São Paulo State University (UNESP), Botucatu 18610-034, SP, Brazil; (C.R.B.); (V.d.A.P.)
| | - Fábio Rodrigues Ferreira Seiva
- Post Graduation Program in Experimental Pathology, State University of Londrina (UEL), Londrina 86057-970, PR, Brazil; (W.L.C.); (M.C.d.S.)
- Department of Chemical and Biological Sciences, Institute of Bioscience, São Paulo State University (UNESP), Botucatu 18610-034, SP, Brazil; (C.R.B.); (V.d.A.P.)
| |
Collapse
|
26
|
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
|
27
|
Ji X, Williams KP, Zheng W. Applying a Gene Reversal Rate Computational Methodology to Identify Drugs for a Rare Cancer: Inflammatory Breast Cancer. Cancer Inform 2023; 22:11769351231202588. [PMID: 37846218 PMCID: PMC10576937 DOI: 10.1177/11769351231202588] [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: 04/11/2023] [Accepted: 09/01/2023] [Indexed: 10/18/2023] Open
Abstract
The aim of this study was to utilize a computational methodology based on Gene Reversal Rate (GRR) scoring to repurpose existing drugs for a rare and understudied cancer: inflammatory breast cancer (IBC). This method uses IBC-related gene expression signatures (GES) and drug-induced gene expression profiles from the LINCS database to calculate a GRR score for each candidate drug, and is based on the idea that a compound that can counteract gene expression changes of a disease may have potential therapeutic applications for that disease. Genes related to IBC with associated differential expression data (265 up-regulated and 122 down-regulated) were collated from PubMed-indexed publications. Drug-induced gene expression profiles were downloaded from the LINCS database and candidate drugs to treat IBC were predicted using their GRR scores. Thirty-two (32) drug perturbations that could potentially reverse the pre-compiled list of 297 IBC genes were obtained using the LINCS Canvas Browser (LCB) analysis. Binary combinations of the 32 perturbations were assessed computationally to identify combined perturbations with the highest GRR scores, and resulted in 131 combinations with GRR greater than 80%, that reverse up to 264 of the 297 genes in the IBC-GES. The top 35 combinations involve 20 unique individual drug perturbations, and 19 potential drug candidates. A comprehensive literature search confirmed 17 of the 19 known drugs as having either anti-cancer or anti-inflammatory activities. AZD-7545, BMS-754807, and nimesulide target known IBC relevant genes: PDK, Met, and COX, respectively. AG-14361, butalbital, and clobenpropit are known to be functionally relevant in DNA damage, cell cycle, and apoptosis, respectively. These findings support the use of the GRR approach to identify drug candidates and potential combination therapies that could be used to treat rare diseases such as IBC.
Collapse
Affiliation(s)
- Xiaojia Ji
- BRITE Institute and Department of Pharmaceutical Sciences, College of Health and Sciences, North Carolina Central University, Durham, NC, USA
| | - Kevin P Williams
- BRITE Institute and Department of Pharmaceutical Sciences, College of Health and Sciences, North Carolina Central University, Durham, NC, USA
| | - Weifan Zheng
- BRITE Institute and Department of Pharmaceutical Sciences, College of Health and Sciences, North Carolina Central University, Durham, NC, USA
| |
Collapse
|
28
|
Tian S, Li Y, Xu J, Zhang L, Zhang J, Lu J, Xu X, Luan X, Zhao J, Zhang W. COIMMR: a computational framework to reveal the contribution of herbal ingredients against human cancer via immune microenvironment and metabolic reprogramming. Brief Bioinform 2023; 24:bbad346. [PMID: 37816138 PMCID: PMC10564268 DOI: 10.1093/bib/bbad346] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/16/2023] [Accepted: 09/13/2023] [Indexed: 10/12/2023] Open
Abstract
Immune evasion and metabolism reprogramming have been regarded as two vital hallmarks of the mechanism of carcinogenesis. Thus, targeting the immune microenvironment and the reprogrammed metabolic processes will aid in developing novel anti-cancer drugs. In recent decades, herbal medicine has been widely utilized to treat cancer through the modulation of the immune microenvironment and reprogrammed metabolic processes. However, labor-based herbal ingredient screening is time consuming, laborious and costly. Luckily, some computational approaches have been proposed to screen candidates for drug discovery rapidly. Yet, it has been challenging to develop methods to screen drug candidates exclusively targeting specific pathways, especially for herbal ingredients which exert anti-cancer effects by multiple targets, multiple pathways and synergistic ways. Meanwhile, currently employed approaches cannot quantify the contribution of the specific pathway to the overall curative effect of herbal ingredients. Hence, to address this problem, this study proposes a new computational framework to infer the contribution of the immune microenvironment and metabolic reprogramming (COIMMR) in herbal ingredients against human cancer and specifically screen herbal ingredients targeting the immune microenvironment and metabolic reprogramming. Finally, COIMMR was applied to identify isoliquiritigenin that specifically regulates the T cells in stomach adenocarcinoma and cephaelin hydrochloride that specifically targets metabolic reprogramming in low-grade glioma. The in silico results were further verified using in vitro experiments. Taken together, our approach opens new possibilities for repositioning drugs targeting immune and metabolic dysfunction in human cancer and provides new insights for drug development in other diseases. COIMMR is available at https://github.com/LYN2323/COIMMR.
Collapse
Affiliation(s)
- Saisai Tian
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Yanan Li
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Jia Xu
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
- College of Pharmacy, Henan University, Kaifeng 475000, China
| | - Lijun Zhang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosafety, Institute of Interdisciplinary Integrative Medicine
| | - Jinbo Zhang
- Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China Department of Pharmacy, Tianjin Rehabilitation Center of Joint Logistics Support Force, Tianjin, 300110, China
| | - Jinyuan Lu
- College of Pharmacy, Anhui University of Chinese Medicine, Anhui 230012, China
| | - Xike Xu
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Xin Luan
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosafety, Institute of Interdisciplinary Integrative Medicine
| | - Jing Zhao
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosafety, Institute of Interdisciplinary Integrative Medicine
| | - Weidong Zhang
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosafety, Institute of Interdisciplinary Integrative Medicine
| |
Collapse
|
29
|
Gusev A. Germline mechanisms of immunotherapy toxicities in the era of genome-wide association studies. Immunol Rev 2023; 318:138-156. [PMID: 37515388 PMCID: PMC11472697 DOI: 10.1111/imr.13253] [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: 03/14/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023]
Abstract
Cancer immunotherapy has revolutionized the treatment of advanced cancers and is quickly becoming an option for early-stage disease. By reactivating the host immune system, immunotherapy harnesses patients' innate defenses to eradicate the tumor. By putatively similar mechanisms, immunotherapy can also substantially increase the risk of toxicities or immune-related adverse events (irAEs). Severe irAEs can lead to hospitalization, treatment discontinuation, lifelong immune complications, or even death. Many irAEs present with similar symptoms to heritable autoimmune diseases, suggesting that germline genetics may contribute to their onset. Recently, genome-wide association studies (GWAS) of irAEs have identified common germline associations and putative mechanisms, lending support to this hypothesis. A wide range of well-established GWAS methods can potentially be harnessed to understand the etiology of irAEs specifically and immunotherapy outcomes broadly. This review summarizes current findings regarding germline effects on immunotherapy outcomes and discusses opportunities and challenges for leveraging germline genetics to understand, predict, and treat irAEs.
Collapse
Affiliation(s)
- Alexander Gusev
- Division of Population Sciences, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics, Brigham & Women's Hospital, Boston, Massachusetts, USA
- The Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
30
|
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
|
31
|
Toh H, Smolentsev A, Sadjadi R, Clegg D, Yan J, Stewart R, Thomson JA, Jiang P. Transcriptomic clock predicts vascular changes of prodromal diabetic retinopathy. Sci Rep 2023; 13:12968. [PMID: 37563287 PMCID: PMC10415264 DOI: 10.1038/s41598-023-40328-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/08/2023] [Indexed: 08/12/2023] Open
Abstract
Diabetic retinopathy is a common complication of long-term diabetes and that could lead to vision loss. Unfortunately, early diabetic retinopathy remains poorly understood. There is no effective way to prevent or treat early diabetic retinopathy until patients develop later stages of diabetic retinopathy. Elevated acellular capillary density is considered a reliable quantitative trait present in the early development of retinopathy. Hence, in this study, we interrogated whole retinal vascular transcriptomic changes via a Nile rat model to better understand the early pathogenesis of diabetic retinopathy. We uncovered the complexity of associations between acellular capillary density and the joint factors of blood glucose, diet, and sex, which was modeled through a Bayesian network. Using segmented regressions, we have identified different gene expression patterns and enriched Gene Ontology (GO) terms associated with acellular capillary density increasing. We developed a random forest regression model based on expression patterns of 14 genes to predict the acellular capillary density. Since acellular capillary density is a reliable quantitative trait in early diabetic retinopathy, and thus our model can be used as a transcriptomic clock to measure the severity of the progression of early retinopathy. We also identified NVP-TAE684, geldanamycin, and NVP-AUY922 as the top three potential drugs which can potentially attenuate the early DR. Although we need more in vivo studies in the future to support our re-purposed drugs, we have provided a data-driven approach to drug discovery.
Collapse
Affiliation(s)
- Huishi Toh
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Alexander Smolentsev
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Ryan Sadjadi
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Dennis Clegg
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Jingqi Yan
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH, 44115, USA
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, 44115, USA
| | - Ron Stewart
- Morgridge Institute For Research, Madison, WI, 53706, USA
| | - James A Thomson
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
- Morgridge Institute For Research, Madison, WI, 53706, USA
| | - Peng Jiang
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, OH, 44115, USA.
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, 44115, USA.
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
32
|
Cüvitoğlu A, Isik Z. Network neighborhood operates as a drug repositioning method for cancer treatment. PeerJ 2023; 11:e15624. [PMID: 37456868 PMCID: PMC10340098 DOI: 10.7717/peerj.15624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 06/01/2023] [Indexed: 07/18/2023] Open
Abstract
Computational drug repositioning approaches are important, as they cost less compared to the traditional drug development processes. This study proposes a novel network-based drug repositioning approach, which computes similarities between disease-causing genes and drug-affected genes in a network topology to suggest candidate drugs with highest similarity scores. This new method aims to identify better treatment options by integrating systems biology approaches. It uses a protein-protein interaction network that is the main topology to compute a similarity score between candidate drugs and disease-causing genes. The disease-causing genes were mapped on this network structure. Transcriptome profiles of drug candidates were taken from the LINCS project and mapped individually on the network structure. The similarity of these two networks was calculated by different network neighborhood metrics, including Adamic-Adar, PageRank and neighborhood scoring. The proposed approach identifies the best candidates by choosing the drugs with significant similarity scores. The method was experimented on melanoma, colorectal, and prostate cancers. Several candidate drugs were predicted by applying AUC values of 0.6 or higher. Some of the predictions were approved by clinical phase trials or other in-vivo studies found in literature. The proposed drug repositioning approach would suggest better treatment options with integration of functional information between genes and transcriptome level effects of drug perturbations and diseases.
Collapse
Affiliation(s)
- Ali Cüvitoğlu
- The Graduate School of Natural and Applied Sciences, Dokuz Eylül University, Izmir, Turkiye
| | - Zerrin Isik
- Computer Engineering Department, Engineering Faculty, Dokuz Eylül University, Izmir, Turkiye
| |
Collapse
|
33
|
Obermayer AN, Chang D, Nobles G, Teng M, Tan AC, Wang X, Chen YA, Eschrich S, Rodriguez PC, Grass GD, Meshinchi S, Tarhini A, Chen DT, Shaw TI. PATH-SURVEYOR: pathway level survival enquiry for immuno-oncology and drug repurposing. BMC Bioinformatics 2023; 24:266. [PMID: 37380943 PMCID: PMC10303868 DOI: 10.1186/s12859-023-05393-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 06/19/2023] [Indexed: 06/30/2023] Open
Abstract
Pathway-level survival analysis offers the opportunity to examine molecular pathways and immune signatures that influence patient outcomes. However, available survival analysis algorithms are limited in pathway-level function and lack a streamlined analytical process. Here we present a comprehensive pathway-level survival analysis suite, PATH-SURVEYOR, which includes a Shiny user interface with extensive features for systematic exploration of pathways and covariates in a Cox proportional-hazard model. Moreover, our framework offers an integrative strategy for performing Hazard Ratio ranked Gene Set Enrichment Analysis and pathway clustering. As an example, we applied our tool in a combined cohort of melanoma patients treated with checkpoint inhibition (ICI) and identified several immune populations and biomarkers predictive of ICI efficacy. We also analyzed gene expression data of pediatric acute myeloid leukemia (AML) and performed an inverse association of drug targets with the patient's clinical endpoint. Our analysis derived several drug targets in high-risk KMT2A-fusion-positive patients, which were then validated in AML cell lines in the Genomics of Drug Sensitivity database. Altogether, the tool offers a comprehensive suite for pathway-level survival analysis and a user interface for exploring drug targets, molecular features, and immune populations at different resolutions.
Collapse
Affiliation(s)
- Alyssa N Obermayer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Darwin Chang
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Gabrielle Nobles
- Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Mingxiang Teng
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Aik-Choon Tan
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Y Ann Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Steven Eschrich
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - G Daniel Grass
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Children's Oncology Group, Monrovia, CA, USA
| | - Ahmad Tarhini
- Department of Cutaneous Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Dung-Tsa Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Timothy I Shaw
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
34
|
Adikusuma W, Zakaria ZA, Irham LM, Nopitasari BL, Pradiningsih A, Firdayani F, Septama AW, Chong R. Transcriptomics-driven drug repositioning for the treatment of diabetic foot ulcer. Sci Rep 2023; 13:10032. [PMID: 37340026 DOI: 10.1038/s41598-023-37120-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/15/2023] [Indexed: 06/22/2023] Open
Abstract
Diabetic foot ulcers (DFUs) are a common complication of diabetes and can lead to severe disability and even amputation. Despite advances in treatment, there is currently no cure for DFUs and available drugs for treatment are limited. This study aimed to identify new candidate drugs and repurpose existing drugs to treat DFUs based on transcriptomics analysis. A total of 31 differentially expressed genes (DEGs) were identified and used to prioritize the biological risk genes for DFUs. Further investigation using the database DGIdb revealed 12 druggable target genes among 50 biological DFU risk genes, corresponding to 31 drugs. Interestingly, we highlighted that two drugs (urokinase and lidocaine) are under clinical investigation for DFU and 29 drugs are potential candidates to be repurposed for DFU therapy. The top 5 potential biomarkers for DFU from our findings are IL6ST, CXCL9, IL1R1, CXCR2, and IL10. This study highlights IL1R1 as a highly promising biomarker for DFU due to its high systemic score in functional annotations, that can be targeted with an existing drug, Anakinra. Our study proposed that the integration of transcriptomic and bioinformatic-based approaches has the potential to drive drug repurposing for DFUs. Further research will further examine the mechanisms by which targeting IL1R1 can be used to treat DFU.
Collapse
Affiliation(s)
- Wirawan Adikusuma
- Borneo Research on Algesia, Inflammation, and Neurodegeneration (BRAIN) Group, Department of Biomedical Sciences, Faculty of Medicines and Health Sciences, University Malaysia Sabah, Jalan UMS, 88400, Kota Kinabalu, Sabah, Malaysia.
- Departement of Pharmacy, University of Muhammadiyah Mataram, Mataram, Indonesia.
- Research Center for Vaccine and Drugs, National Research and Innovation Agency (BRIN), South Tangerang, Indonesia.
| | - Zainul Amiruddin Zakaria
- Borneo Research on Algesia, Inflammation, and Neurodegeneration (BRAIN) Group, Department of Biomedical Sciences, Faculty of Medicines and Health Sciences, University Malaysia Sabah, Jalan UMS, 88400, Kota Kinabalu, Sabah, Malaysia.
| | - Lalu Muhammad Irham
- Faculty of Pharmacy, Universitas Ahmad Dahlan, Yogyakarta, Indonesia
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), South Tangerang, Indonesia
| | | | - Anna Pradiningsih
- Departement of Pharmacy, University of Muhammadiyah Mataram, Mataram, Indonesia
| | - Firdayani Firdayani
- Research Center for Vaccine and Drugs, National Research and Innovation Agency (BRIN), South Tangerang, Indonesia
| | - Abdi Wira Septama
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), South Tangerang, Indonesia
| | - Rockie Chong
- Department of Chemistry and Biochemistry, University of California, Los Angeles, USA
| |
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
|
Gopalan V, Hannenhalli S. Towards a Synthesis of the Non-Genetic and Genetic Views of Cancer in Understanding Pancreatic Ductal Adenocarcinoma Initiation and Prevention. Cancers (Basel) 2023; 15:cancers15072159. [PMID: 37046820 PMCID: PMC10093726 DOI: 10.3390/cancers15072159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
While much of the research in oncogenesis and cancer therapy has focused on mutations in key cancer driver genes, more recent work suggests a complementary non-genetic paradigm. This paradigm focuses on how transcriptional and phenotypic heterogeneity, even in clonally derived cells, can create sub-populations associated with oncogenesis, metastasis, and therapy resistance. We discuss this complementary paradigm in the context of pancreatic ductal adenocarcinoma. A better understanding of cellular transcriptional heterogeneity and its association with oncogenesis can lead to more effective therapies that prevent tumor initiation and slow progression.
Collapse
Affiliation(s)
- Vishaka Gopalan
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| |
Collapse
|
37
|
Groenewald W, Lund AH, Gay DM. The Role of WNT Pathway Mutations in Cancer Development and an Overview of Therapeutic Options. Cells 2023; 12:990. [PMID: 37048063 PMCID: PMC10093220 DOI: 10.3390/cells12070990] [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: 02/27/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 04/14/2023] Open
Abstract
It is well established that mutations in the canonical WNT-signalling pathway play a major role in various cancers. Critical to developing new therapeutic strategies is understanding which cancers are driven by WNT pathway activation and at what level these mutations occur within the pathway. Some cancers harbour mutations in genes whose protein products operate at the receptor level of the WNT pathway. For instance, tumours with RNF43 or RSPO mutations, still require exogenous WNT ligands to drive WNT signalling (ligand-dependent mutations). Conversely, mutations within the cytoplasmic segment of the Wnt pathway, such as in APC and CTNNB1, lead to constitutive WNT pathway activation even in the absence of WNT ligands (ligand-independent). Here, we review the predominant driving mutations found in cancer that lead to WNT pathway activation, as well as explore some of the therapeutic interventions currently available against tumours harbouring either ligand-dependent or ligand-independent mutations. Finally, we discuss a potentially new therapeutic avenue by targeting the translational apparatus downstream from WNT signalling.
Collapse
Affiliation(s)
| | - Anders H. Lund
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - David Michael Gay
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
38
|
Foo RJK, Tian S, Tan EY, Goh WWB. A novel survival prediction signature outperforms PAM50 and artificial intelligence-based feature-selection methods. Comput Biol Chem 2023; 104:107845. [PMID: 36889140 DOI: 10.1016/j.compbiolchem.2023.107845] [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: 09/09/2022] [Revised: 01/06/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023]
Abstract
The robustness of a breast cancer gene signature, the super-proliferation set (SPS), is initially tested and investigated on breast cancer cell lines from the Cancer Cell Line Encyclopaedia (CCLE). Previously, SPS was derived via a meta-analysis of 47 independent breast cancer gene signatures, benchmarked on survival information from clinical data in the NKI dataset. Here, relying on the stability of cell line data and associative prior knowledge, we first demonstrate through Principal Component Analysis (PCA) that SPS prioritizes survival information over secondary subtype information, surpassing both PAM50 and Boruta, an artificial intelligence-based feature-selection algorithm, in this regard. We can also extract higher resolution 'progression' information using SPS, dividing survival outcomes into several clinically relevant stages ('good', 'intermediate', and 'bad) based on different quadrants of the PCA scatterplot. Furthermore, by transferring these 'progression' annotations onto independent clinical datasets, we demonstrate the generalisability of our method on actual patient data. Finally, via the characteristic genetic profiles of each quadrant/stage, we identified efficacious drugs using their gene reversal scores that can shift signatures across quadrants/stages, in a process known as gene signature reversal. This confirms the power of meta-analytical approaches for gene signature inference in breast cancer, as well as the clinical benefit in translating these inferences onto real-world patient data for more targeted therapies.
Collapse
Affiliation(s)
- Reuben Jyong Kiat Foo
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Siqi Tian
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore
| | - Ern Yu Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Tan Tock Seng Hospital, Singapore
| | - Wilson Wen Bin Goh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore; Centre for Biomedical Informatics, Nanyang Technological University, Singapore.
| |
Collapse
|
39
|
Torricelli F, Sauta E, Manicardi V, Mandato VD, Palicelli A, Ciarrocchi A, Manzotti G. An Innovative Drug Repurposing Approach to Restrain Endometrial Cancer Metastatization. Cells 2023; 12:794. [PMID: 36899930 PMCID: PMC10001006 DOI: 10.3390/cells12050794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Endometrial cancer (EC) is the most common gynecologic tumor and the world's fourth most common cancer in women. Most patients respond to first-line treatments and have a low risk of recurrence, but refractory patients, and those with metastatic cancer at diagnosis, remain with no treatment options. Drug repurposing aims to discover new clinical indications for existing drugs with known safety profiles. It provides ready-to-use new therapeutic options for highly aggressive tumors for which standard protocols are ineffective, such as high-risk EC. METHODS Here, we aimed at defining new therapeutic opportunities for high-risk EC using an innovative and integrated computational drug repurposing approach. RESULTS We compared gene-expression profiles, from publicly available databases, of metastatic and non-metastatic EC patients being metastatization the most severe feature of EC aggressiveness. A comprehensive analysis of transcriptomic data through a two-arm approach was applied to obtain a robust prediction of drug candidates. CONCLUSIONS Some of the identified therapeutic agents are already successfully used in clinical practice to treat other types of tumors. This highlights the potential to repurpose them for EC and, therefore, the reliability of the proposed approach.
Collapse
Affiliation(s)
- Federica Torricelli
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - Elisabetta Sauta
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, 27100 Pavia, Italy
| | - Veronica Manicardi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, 27100 Pavia, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Vincenzo Dario Mandato
- Unit of Obstetrics and Gynaecology, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Andrea Palicelli
- Pathology Unit, Department of Oncology and Advanced Technologies, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - Gloria Manzotti
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| |
Collapse
|
40
|
Liu Y, Song F, Li Z, Chen L, Xu Y, Sun H, Chang Y. A comprehensive tool for tumor precision medicine with pharmaco-omics data analysis. Front Pharmacol 2023; 14:1085765. [PMID: 36713829 PMCID: PMC9878337 DOI: 10.3389/fphar.2023.1085765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/04/2023] [Indexed: 01/14/2023] Open
Abstract
Background: Cancer precision medicine is an effective strategy to fight cancers by bridging genomics and drug discovery to provide specific treatment for patients with different genetic characteristics. Although some public databases and modelling frameworks have been developed through studies on drug response, most of them only considered the ramifications of the drug on the cell line and the effects on the patient still require a huge amount of work to integrate data from various databases and calculations, especially concerning precision treatment. Furthermore, not only efficacy but also the adverse effects of drugs on patients should be taken into account during cancer treatment. However, the adverse effects as essential indicators of drug safety assessment are always neglected. Method: A holistic estimation explores various drugs' efficacy levels by calculating their potency both in reversing and enhancing cancer-associated gene expression change. And a method for bridging the gap between cell culture and living tissue estimates the effectiveness of a drug on individual patients through the mappings of various cell lines to each person according to their genetic mutation similarities. Result: We predicted the efficacy of FDA-recommended drugs, taking into account both efficacy and toxicity, and obtained consistent results. We also provided an intuitive and easy-to-use web server called DBPOM (http://www.dbpom.net/, a comprehensive database of pharmaco-omics for cancer precision medicine), which not only integrates the above methods but also provides calculation results on more than 10,000 small molecule compounds and drugs. As a one-stop web server, clinicians and drug researchers can also analyze the overall effect of a drug or a drug combination on cancer patients as well as the biological functions that they target. DBPOM is now public, free to use with no login requirement, and contains all the data and code. Conclusion: Both the positive and negative effects of drugs during precision treatment are essential for practical application of drugs. DBPOM based on the two effects will become a vital resource and analysis platform for drug development, drug mechanism studies and the discovery of new therapies.
Collapse
Affiliation(s)
- Yijun Liu
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Fuhu Song
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Zhi Li
- Medical Oncology Department, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Liang Chen
- Department of Computer Science, College of Engineering, Shantou University, Shantou, China,Key Laboratory of Intelligent Manufacturing Technology of Ministry of Education, Shantou University, Shantou, China
| | - Ying Xu
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology, Institute of Bioinformatics, The University of Georgia, Athens, GA, United States
| | - Huiyan Sun
- School of Artificial Intelligence, Jilin University, Changchun, China,International Center of Future Science, Jilin University, Changchun, China,*Correspondence: Huiyan Sun, ; Yi Chang,
| | - Yi Chang
- School of Artificial Intelligence, Jilin University, Changchun, China,International Center of Future Science, Jilin University, Changchun, China,*Correspondence: Huiyan Sun, ; Yi Chang,
| |
Collapse
|
41
|
Bruggemann L, Falls Z, Mangione W, Schwartz SA, Battaglia S, Aalinkeel R, Mahajan SD, Samudrala R. Multiscale Analysis and Validation of Effective Drug Combinations Targeting Driver KRAS Mutations in Non-Small Cell Lung Cancer. Int J Mol Sci 2023; 24:ijms24020997. [PMID: 36674513 PMCID: PMC9867122 DOI: 10.3390/ijms24020997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 01/06/2023] Open
Abstract
Pharmacogenomics is a rapidly growing field with the goal of providing personalized care to every patient. Previously, we developed the Computational Analysis of Novel Drug Opportunities (CANDO) platform for multiscale therapeutic discovery to screen optimal compounds for any indication/disease by performing analytics on their interactions using large protein libraries. We implemented a comprehensive precision medicine drug discovery pipeline within the CANDO platform to determine which drugs are most likely to be effective against mutant phenotypes of non-small cell lung cancer (NSCLC) based on the supposition that drugs with similar interaction profiles (or signatures) will have similar behavior and therefore show synergistic effects. CANDO predicted that osimertinib, an EGFR inhibitor, is most likely to synergize with four KRAS inhibitors.Validation studies with cellular toxicity assays confirmed that osimertinib in combination with ARS-1620, a KRAS G12C inhibitor, and BAY-293, a pan-KRAS inhibitor, showed a synergistic effect on decreasing cellular proliferation by acting on mutant KRAS. Gene expression studies revealed that MAPK expression is strongly correlated with decreased cellular proliferation following treatment with KRAS inhibitor BAY-293, but not treatment with ARS-1620 or osimertinib. These results indicate that our precision medicine pipeline may be used to identify compounds capable of synergizing with inhibitors of KRAS G12C, and to assess their likelihood of becoming drugs by understanding their behavior at the proteomic/interactomic scales.
Collapse
Affiliation(s)
- Liana Bruggemann
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY 14260, USA
| | - Zackary Falls
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY 14260, USA
| | - William Mangione
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY 14260, USA
| | | | | | | | - Supriya D. Mahajan
- Department of Medicine, University at Buffalo, Buffalo, NY 14260, USA
- Correspondence: (S.D.M.); (R.S.)
| | - Ram Samudrala
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY 14260, USA
- Correspondence: (S.D.M.); (R.S.)
| |
Collapse
|
42
|
Papikinos T, Krokidis MG, Vrahatis A, Vlamos P, Exarchos TP. Signature-Based Computational Drug Repurposing for Amyotrophic Lateral Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1424:201-211. [PMID: 37486495 DOI: 10.1007/978-3-031-31982-2_22] [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: 07/25/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a late-onset fatal neurodegenerative disease characterized by progressive loss of the upper and lower motor neurons. There are currently limited approved drugs for the disorder, and for this reason the strategy of repositioning already approved therapeutics could exhibit a successful outcome. Herein, we used CMAP and L1000CDS2 databases which include gene expression profiles datasets (genomic signatures) to identify potent compounds and classes of compounds which reverse disease's signature which could in turn reverse its phenotype. ALS signature was obtained by comparing gene expression of muscle biopsy specimens between diseased and healthy individuals. Statistical analysis was conducted to explore differentially transcripts in patients' samples. Then, the list of upregulated and downregulated genes was used to query both databases in order to determine molecules which downregulate the genes which are upregulated by ALS and vice versa. These compounds, based on their chemical structure along with known treatments, were clustered to reveal drugs with novel and potentially more effective mode of action with most of them predicted to affect pathways heavily involved in ALS. This evidence suggests that these compounds are strong candidates for moving to the next phase of the drug repurposing pipeline which is in vitro and in vivo experimental evaluation.
Collapse
Affiliation(s)
- Thomas Papikinos
- Department of Informatics, Bioinformatics and Human Electrophysiology Laboratory, Ionian University, Corfu, Greece.
| | - Marios G Krokidis
- Department of Informatics, Bioinformatics and Human Electrophysiology Laboratory, Ionian University, Corfu, Greece
| | - Aris Vrahatis
- Department of Informatics, Bioinformatics and Human Electrophysiology Laboratory, Ionian University, Corfu, Greece
| | - Panagiotis Vlamos
- Department of Informatics, Bioinformatics and Human Electrophysiology Laboratory, Ionian University, Corfu, Greece
| | - Themis P Exarchos
- Department of Informatics, Bioinformatics and Human Electrophysiology Laboratory, Ionian University, Corfu, Greece
| |
Collapse
|
43
|
Ko M, Oh JM, Kim IW. Drug repositioning prediction for psoriasis using the adverse event reporting database. Front Med (Lausanne) 2023; 10:1159453. [PMID: 37035327 PMCID: PMC10076533 DOI: 10.3389/fmed.2023.1159453] [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: 02/06/2023] [Accepted: 02/28/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Inverse signals produced from disproportional analyses using spontaneous drug adverse event reports can be used for drug repositioning purposes. The purpose of this study is to predict drug candidates using a computational method that integrates reported drug adverse event data, disease-specific gene expression profiles, and drug-induced gene expression profiles. Methods Drug and adverse events from 2015 through 2020 were downloaded from the United States Food and Drug Administration Adverse Event Reporting System (FAERS). The reporting odds ratio (ROR), information component (IC) and empirical Bayes geometric mean (EBGM) were used to calculate the inverse signals. Psoriasis was selected as the target disease. Disease specific gene expression profiles were obtained by the meta-analysis of the Gene Expression Omnibus (GEO). The reverse gene expression scores were calculated using the Library of Integrated Network-based Cellular Signatures (LINCS) and their correlations with the inverse signals were obtained. Results Reversal genes and the candidate compounds were identified. Additionally, these correlations were validated using the relationship between the reverse gene expression scores and the half-maximal inhibitory concentration (IC50) values from the Chemical European Molecular Biology Laboratory (ChEMBL). Conclusion Inverse signals produced from a disproportional analysis can be used for drug repositioning and to predict drug candidates against psoriasis.
Collapse
Affiliation(s)
- Minoh Ko
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Jung Mi Oh
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - In-Wha Kim
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
- *Correspondence: In-Wha Kim,
| |
Collapse
|
44
|
Koudijs KKM, Böhringer S, Guchelaar HJ. Validation of transcriptome signature reversion for drug repurposing in oncology. Brief Bioinform 2022; 24:6850563. [PMID: 36445193 PMCID: PMC9851289 DOI: 10.1093/bib/bbac490] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/21/2022] [Accepted: 10/15/2022] [Indexed: 11/30/2022] Open
Abstract
Transcriptome signature reversion (TSR) has been extensively proposed and used to discover new indications for existing drugs (i.e. drug repositioning, drug repurposing) for various cancer types. TSR relies on the assumption that a drug that can revert gene expression changes induced by a disease back to original, i.e. healthy, levels is likely to be therapeutically active in treating the disease. Here, we aimed to validate the concept of TSR using the PRISM repurposing data set, which is-as of writing-the largest pharmacogenomic data set. The predictive utility of the TSR approach as it has currently been used appears to be much lower than previously reported and is completely nullified after the drug gene expression signatures are adjusted for the general anti-proliferative downstream effects of drug-induced decreased cell viability. Therefore, TSR mainly relies on generic anti-proliferative drug effects rather than on targeting cancer pathways specifically upregulated in tumor types.
Collapse
Affiliation(s)
- Karel K M Koudijs
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center (LUMC); 2333 ZA Leiden, The Netherlands
| | - Stefan Böhringer
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center (LUMC); 2333 ZA Leiden, The Netherlands,Department of Biomedical Data Sciences, Leiden University Medical Center (LUMC); 2333 ZA Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Corresponding author: Henk-Jan Guchelaar, Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands. Tel.: +31-71-526-4018; Fax: +31-71-526-6980; E-mail:
| |
Collapse
|
45
|
Singha M, Pu L, Stanfield BA, Uche IK, Rider PJF, Kousoulas KG, Ramanujam J, Brylinski M. Artificial intelligence to guide precision anticancer therapy with multitargeted kinase inhibitors. BMC Cancer 2022; 22:1211. [PMID: 36434556 PMCID: PMC9694576 DOI: 10.1186/s12885-022-10293-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 11/07/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Vast amounts of rapidly accumulating biological data related to cancer and a remarkable progress in the field of artificial intelligence (AI) have paved the way for precision oncology. Our recent contribution to this area of research is CancerOmicsNet, an AI-based system to predict the therapeutic effects of multitargeted kinase inhibitors across various cancers. This approach was previously demonstrated to outperform other deep learning methods, graph kernel models, molecular docking, and drug binding pocket matching. METHODS CancerOmicsNet integrates multiple heterogeneous data by utilizing a deep graph learning model with sophisticated attention propagation mechanisms to extract highly predictive features from cancer-specific networks. The AI-based system was devised to provide more accurate and robust predictions than data-driven therapeutic discovery using gene signature reversion. RESULTS Selected CancerOmicsNet predictions obtained for "unseen" data are positively validated against the biomedical literature and by live-cell time course inhibition assays performed against breast, pancreatic, and prostate cancer cell lines. Encouragingly, six molecules exhibited dose-dependent antiproliferative activities, with pan-CDK inhibitor JNJ-7706621 and Src inhibitor PP1 being the most potent against the pancreatic cancer cell line Panc 04.03. CONCLUSIONS CancerOmicsNet is a promising AI-based platform to help guide the development of new approaches in precision oncology involving a variety of tumor types and therapeutics.
Collapse
Affiliation(s)
- Manali Singha
- grid.64337.350000 0001 0662 7451Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Limeng Pu
- grid.64337.350000 0001 0662 7451Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Brent A. Stanfield
- grid.64337.350000 0001 0662 7451Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Ifeanyi K. Uche
- grid.64337.350000 0001 0662 7451Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.64337.350000 0001 0662 7451Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.279863.10000 0000 8954 1233School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112 USA
| | - Paul J. F. Rider
- grid.64337.350000 0001 0662 7451Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.64337.350000 0001 0662 7451Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Konstantin G. Kousoulas
- grid.64337.350000 0001 0662 7451Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.64337.350000 0001 0662 7451Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA
| | - J. Ramanujam
- grid.64337.350000 0001 0662 7451Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.64337.350000 0001 0662 7451Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Michal Brylinski
- grid.64337.350000 0001 0662 7451Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.64337.350000 0001 0662 7451Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803 USA
| |
Collapse
|
46
|
Jang HY, Oh JM, Kim IW. Drug repurposing using meta-analysis of gene expression in Alzheimer's disease. Front Neurosci 2022; 16:989174. [PMID: 36440278 PMCID: PMC9684643 DOI: 10.3389/fnins.2022.989174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/19/2022] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION Alzheimer's disease and other forms of dementia are disease that bring an increased global burden. However, the medicine developed to date remains limited. The purpose of this study is to predict drug repositioning candidates using a computational method that integrates gene expression profiles on Alzheimer's disease and compound-induced changes in gene expression levels. METHODS Gene expression data on Alzheimer's disease were obtained from the Gene Expression Omnibus (GEO) and we conducted a meta-analysis of their gene expression levels. The reverse scores of compound-induced gene expressions were computed based on the reversal relationship between disease and drug gene expression profiles. RESULTS Reversal genes and the candidate compounds were identified by the leave-one-out cross-validation procedure. Additionally, the half-maximal inhibitory concentration (IC50) values and the blood-brain barrier (BBB) permeability of candidate compounds were obtained from ChEMBL and PubChem, respectively. CONCLUSION New therapeutic target genes and drug candidates against Alzheimer's disease were identified by means of drug repositioning.
Collapse
Affiliation(s)
- Ha Young Jang
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Jung Mi Oh
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea,College of Pharmacy, Seoul National University, Seoul, South Korea
| | - In-Wha Kim
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea,*Correspondence: In-Wha Kim,
| |
Collapse
|
47
|
Jiménez‐Santos MJ, García‐Martín S, Fustero‐Torre C, Di Domenico T, Gómez‐López G, Al‐Shahrour F. Bioinformatics roadmap for therapy selection in cancer genomics. Mol Oncol 2022; 16:3881-3908. [PMID: 35811332 PMCID: PMC9627786 DOI: 10.1002/1878-0261.13286] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/22/2022] [Accepted: 07/08/2022] [Indexed: 12/24/2022] Open
Abstract
Tumour heterogeneity is one of the main characteristics of cancer and can be categorised into inter- or intratumour heterogeneity. This heterogeneity has been revealed as one of the key causes of treatment failure and relapse. Precision oncology is an emerging field that seeks to design tailored treatments for each cancer patient according to epidemiological, clinical and omics data. This discipline relies on bioinformatics tools designed to compute scores to prioritise available drugs, with the aim of helping clinicians in treatment selection. In this review, we describe the current approaches for therapy selection depending on which type of tumour heterogeneity is being targeted and the available next-generation sequencing data. We cover intertumour heterogeneity studies and individual treatment selection using genomics variants, expression data or multi-omics strategies. We also describe intratumour dissection through clonal inference and single-cell transcriptomics, in each case providing bioinformatics tools for tailored treatment selection. Finally, we discuss how these therapy selection workflows could be integrated into the clinical practice.
Collapse
Affiliation(s)
| | | | - Coral Fustero‐Torre
- Bioinformatics UnitSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Tomás Di Domenico
- Bioinformatics UnitSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Gonzalo Gómez‐López
- Bioinformatics UnitSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Fátima Al‐Shahrour
- Bioinformatics UnitSpanish National Cancer Research Centre (CNIO)MadridSpain
| |
Collapse
|
48
|
Zhao G, Newbury P, Ishi Y, Chekalin E, Zeng B, Glicksberg BS, Wen A, Paithankar S, Sasaki T, Suri A, Nazarian J, Pacold ME, Brat DJ, Nicolaides T, Chen B, Hashizume R. Reversal of cancer gene expression identifies repurposed drugs for diffuse intrinsic pontine glioma. Acta Neuropathol Commun 2022; 10:150. [PMID: 36274161 PMCID: PMC9590174 DOI: 10.1186/s40478-022-01463-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 10/13/2022] [Indexed: 11/25/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is an aggressive incurable brainstem tumor that targets young children. Complete resection is not possible, and chemotherapy and radiotherapy are currently only palliative. This study aimed to identify potential therapeutic agents using a computational pipeline to perform an in silico screen for novel drugs. We then tested the identified drugs against a panel of patient-derived DIPG cell lines. Using a systematic computational approach with publicly available databases of gene signature in DIPG patients and cancer cell lines treated with a library of clinically available drugs, we identified drug hits with the ability to reverse a DIPG gene signature to one that matches normal tissue background. The biological and molecular effects of drug treatment was analyzed by cell viability assay and RNA sequence. In vivo DIPG mouse model survival studies were also conducted. As a result, two of three identified drugs showed potency against the DIPG cell lines Triptolide and mycophenolate mofetil (MMF) demonstrated significant inhibition of cell viability in DIPG cell lines. Guanosine rescued reduced cell viability induced by MMF. In vivo, MMF treatment significantly inhibited tumor growth in subcutaneous xenograft mice models. In conclusion, we identified clinically available drugs with the ability to reverse DIPG gene signatures and anti-DIPG activity in vitro and in vivo. This novel approach can repurpose drugs and significantly decrease the cost and time normally required in drug discovery.
Collapse
Affiliation(s)
- Guisheng Zhao
- grid.137628.90000 0004 1936 8753Department of Pediatrics, New York University Langone Health, 160 East 32nd St., New York, NY 10016 USA
| | - Patrick Newbury
- grid.17088.360000 0001 2150 1785Department of Pediatrics and Human Development, Michigan State University, Secchia Center, Room 732, 15 Michigan St. NE, Grand Rapids, MI 49503 USA
| | - Yukitomo Ishi
- grid.16753.360000 0001 2299 3507Department of Pediatrics, Northwestern University Feinberg School of Medicine, 303 East Superior St., Simpson Querrey 4-514, Chicago, IL 60611 USA ,grid.413808.60000 0004 0388 2248Division of Hematology, Oncology, Neuro-Oncology & Stem Cell Transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 East Chicago Avenue, Box 205, Chicago, IL 60611 USA
| | - Eugene Chekalin
- grid.17088.360000 0001 2150 1785Department of Pediatrics and Human Development, Michigan State University, Secchia Center, Room 732, 15 Michigan St. NE, Grand Rapids, MI 49503 USA
| | - Billy Zeng
- grid.17088.360000 0001 2150 1785Department of Pediatrics and Human Development, Michigan State University, Secchia Center, Room 732, 15 Michigan St. NE, Grand Rapids, MI 49503 USA
| | - Benjamin S. Glicksberg
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029 USA ,grid.416167.30000 0004 0442 1996Icahn School of Medicine at Mount Sinai, Hasso Plattner Institute for Digital Health at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029 USA
| | - Anita Wen
- grid.17088.360000 0001 2150 1785Department of Pediatrics and Human Development, Michigan State University, Secchia Center, Room 732, 15 Michigan St. NE, Grand Rapids, MI 49503 USA
| | - Shreya Paithankar
- grid.17088.360000 0001 2150 1785Department of Pediatrics and Human Development, Michigan State University, Secchia Center, Room 732, 15 Michigan St. NE, Grand Rapids, MI 49503 USA
| | - Takahiro Sasaki
- grid.16753.360000 0001 2299 3507Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, 303 East Superior St., Chicago, IL 60611 USA ,grid.412857.d0000 0004 1763 1087Department of Neurological Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Japan
| | - Amreena Suri
- grid.16753.360000 0001 2299 3507Department of Pediatrics, Northwestern University Feinberg School of Medicine, 303 East Superior St., Simpson Querrey 4-514, Chicago, IL 60611 USA ,grid.413808.60000 0004 0388 2248Division of Hematology, Oncology, Neuro-Oncology & Stem Cell Transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 East Chicago Avenue, Box 205, Chicago, IL 60611 USA
| | - Javad Nazarian
- grid.239560.b0000 0004 0482 1586Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA ,grid.412341.10000 0001 0726 4330University Children’s Hospital Zurich, Steinwiesstrasse 75, 8032 Zurich, Switzerland
| | - Michael E. Pacold
- grid.137628.90000 0004 1936 8753Department of Radiation Oncology, New York University Langone Health, 550 First Avenue, New York, NY 10016 USA
| | - Daniel J. Brat
- grid.16753.360000 0001 2299 3507Department of Pathology, Robert H. Lurie Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave., Chicago, IL 60611 USA
| | - Theodore Nicolaides
- grid.137628.90000 0004 1936 8753Department of Pediatrics, New York University Langone Health, 160 East 32nd St., New York, NY 10016 USA
| | - Bin Chen
- Department of Pediatrics and Human Development, Michigan State University, Secchia Center, Room 732, 15 Michigan St. NE, Grand Rapids, MI, 49503, USA. .,Department of Pharmacology and Toxicology, Michigan State University, 1355 Bogue St, East Lansing, MI, 48824, USA. .,Department of Computer Science and Engineering, Michigan State University, 428 S. Shaw Lane, East Lansing, MI, 48824, USA.
| | - Rintaro Hashizume
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, 303 East Superior St., Simpson Querrey 4-514, Chicago, IL, 60611, USA. .,Division of Hematology, Oncology, Neuro-Oncology & Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 East Chicago Avenue, Box 205, Chicago, IL, 60611, USA. .,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 East Superior St., Chicago, IL, 60611, USA.
| |
Collapse
|
49
|
Xing J, Shankar R, Ko M, Zhang K, Zhang S, Drelich A, Paithankar S, Chekalin E, Chua MS, Rajasekaran S, Kent Tseng CT, Zheng M, Kim S, Chen B. Deciphering COVID-19 host transcriptomic complexity and variations for therapeutic discovery against new variants. iScience 2022; 25:105068. [PMID: 36093376 PMCID: PMC9439871 DOI: 10.1016/j.isci.2022.105068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/11/2022] [Accepted: 08/30/2022] [Indexed: 12/04/2022] Open
Abstract
The molecular manifestations of host cells responding to SARS-CoV-2 and its evolving variants of infection are vastly different across the studied models and conditions, imposing challenges for host-based antiviral drug discovery. Based on the postulation that antiviral drugs tend to reverse the global host gene expression induced by viral infection, we retrospectively evaluated hundreds of signatures derived from 1,700 published host transcriptomic profiles of SARS/MERS/SARS-CoV-2 infection using an iterative data-driven approach. A few of these signatures could be reversed by known anti-SARS-CoV-2 inhibitors, suggesting the potential of extrapolating the biology for new variant research. We discovered IMD-0354 as a promising candidate to reverse the signatures globally with nanomolar IC50 against SARS-CoV-2 and its five variants. IMD-0354 stimulated type I interferon antiviral response, inhibited viral entry, and down-regulated hijacked proteins. This study demonstrates that the conserved coronavirus signatures and the transcriptomic reversal approach that leverages polypharmacological effects could guide new variant therapeutic discovery.
Collapse
Affiliation(s)
- Jing Xing
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI 49503, USA
| | - Rama Shankar
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI 49503, USA
| | - Meehyun Ko
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, 13488, Korea
| | - Keke Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Sulin Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Aleksandra Drelich
- Departments of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Shreya Paithankar
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI 49503, USA
| | - Eugene Chekalin
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI 49503, USA
| | - Mei-Sze Chua
- Department of Surgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Surender Rajasekaran
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI 49503, USA
- Helen DeVos Children’s Hospital, Grand Rapids, MI 49503, USA
| | - Chien-Te Kent Tseng
- Departments of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA
- Center of Biodefense and Emerging Infectious Diseases, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Seungtaek Kim
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, 13488, Korea
| | - Bin Chen
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Computer Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
50
|
Kakoti BB, Bezbaruah R, Ahmed N. Therapeutic drug repositioning with special emphasis on neurodegenerative diseases: Threats and issues. Front Pharmacol 2022; 13:1007315. [PMID: 36263141 PMCID: PMC9574100 DOI: 10.3389/fphar.2022.1007315] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/12/2022] [Indexed: 11/21/2022] Open
Abstract
Drug repositioning or repurposing is the process of discovering leading-edge indications for authorized or declined/abandoned molecules for use in different diseases. This approach revitalizes the traditional drug discovery method by revealing new therapeutic applications for existing drugs. There are numerous studies available that highlight the triumph of several drugs as repurposed therapeutics. For example, sildenafil to aspirin, thalidomide to adalimumab, and so on. Millions of people worldwide are affected by neurodegenerative diseases. According to a 2021 report, the Alzheimer's disease Association estimates that 6.2 million Americans are detected with Alzheimer's disease. By 2030, approximately 1.2 million people in the United States possibly acquire Parkinson's disease. Drugs that act on a single molecular target benefit people suffering from neurodegenerative diseases. Current pharmacological approaches, on the other hand, are constrained in their capacity to unquestionably alter the course of the disease and provide patients with inadequate and momentary benefits. Drug repositioning-based approaches appear to be very pertinent, expense- and time-reducing strategies for the enhancement of medicinal opportunities for such diseases in the current era. Kinase inhibitors, for example, which were developed for various oncology indications, demonstrated significant neuroprotective effects in neurodegenerative diseases. This review expounds on the classical and recent examples of drug repositioning at various stages of drug development, with a special focus on neurodegenerative disorders and the aspects of threats and issues viz. the regulatory, scientific, and economic aspects.
Collapse
Affiliation(s)
- Bibhuti Bhusan Kakoti
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, India
| | | | | |
Collapse
|