1
|
Fillinger L, Walter S, Ley M, Kęska-Izworska K, Dehkordi LG, Kratochwill K, Perco P. Computational modeling approaches and regulatory pathways for drug combinations. Drug Discov Today 2025; 30:104345. [PMID: 40158836 DOI: 10.1016/j.drudis.2025.104345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/28/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Drug combinations offer several advantages over monotherapies, but identifying effective drug combinations while avoiding adverse effects is a major challenge. Computational network models are particularly useful for identifying mechanistically compatible drug combinations and generating hypotheses about their mechanisms of action. Here, we discuss the advantages and challenges of in silico discovery approaches for drug combinations. Obtaining regulatory approval during later stages of product development can be more complex for drug combinations than for single drugs. The regulatory pathway is mainly determined by the approval status of the individual compounds included in a combination. We provide an overview of the regulatory guidelines for drug combination development and discuss trends from previous approvals.
Collapse
Affiliation(s)
| | | | - Matthias Ley
- Delta4 GmbH, Vienna, Austria; Medical University of Vienna, Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Nephrology and Gastroenterology, Vienna, Austria
| | | | | | - Klaus Kratochwill
- Delta4 GmbH, Vienna, Austria; Medical University of Vienna, Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Nephrology and Gastroenterology, Vienna, Austria
| | - Paul Perco
- Delta4 GmbH, Vienna, Austria; Medical University of Innsbruck, Department of Internal Medicine IV, Innsbruck, Austria.
| |
Collapse
|
2
|
Jung S. Advances in modeling cellular state dynamics: integrating omics data and predictive techniques. Anim Cells Syst (Seoul) 2025; 29:72-83. [PMID: 39807350 PMCID: PMC11727055 DOI: 10.1080/19768354.2024.2449518] [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: 10/15/2024] [Revised: 12/19/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025] Open
Abstract
Dynamic modeling of cellular states has emerged as a pivotal approach for understanding complex biological processes such as cell differentiation, disease progression, and tissue development. This review provides a comprehensive overview of current approaches for modeling cellular state dynamics, focusing on techniques ranging from dynamic or static biomolecular network models to deep learning models. We highlight how these approaches integrated with various omics data such as transcriptomics, and single-cell RNA sequencing could be used to capture and predict cellular behavior and transitions. We also discuss applications of these modeling approaches in predicting gene knockout effects, designing targeted interventions, and simulating organ development. This review emphasizes the importance of selecting appropriate modeling strategies based on scalability and resolution requirements, which vary according to the complexity and size of biological systems under study. By evaluating strengths, limitations, and recent advancements of these methodologies, we aim to guide future research in developing more robust and interpretable models for understanding and manipulating cellular state dynamics in various biological contexts, ultimately advancing therapeutic strategies and precision medicine.
Collapse
Affiliation(s)
- Sungwon Jung
- Department of Genome Medicine and Science, Gachon University College of Medicine, Incheon, Republic of Korea
- Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, Republic of Korea
| |
Collapse
|
3
|
Fan K, Gökbağ B, Tang S, Li S, Huang Y, Wang L, Cheng L, Li L. Synthetic lethal connectivity and graph transformer improve synthetic lethality prediction. Brief Bioinform 2024; 25:bbae425. [PMID: 39210507 PMCID: PMC11361842 DOI: 10.1093/bib/bbae425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/14/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Synthetic lethality (SL) has shown great promise for the discovery of novel targets in cancer. CRISPR double-knockout (CDKO) technologies can only screen several hundred genes and their combinations, but not genome-wide. Therefore, good SL prediction models are highly needed for genes and gene pairs selection in CDKO experiments. However, lack of scalable SL properties prevents generalizability of SL interactions to out-of-sample data, thereby hindering modeling efforts. In this paper, we recognize that SL connectivity is a scalable and generalizable SL property. We develop a novel two-step multilayer encoder for individual sample-specific SL prediction model (MLEC-iSL), which predicts SL connectivity first and SL interactions subsequently. MLEC-iSL has three encoders, namely, gene, graph, and transformer encoders. MLEC-iSL achieves high SL prediction performance in K562 (AUPR, 0.73; AUC, 0.72) and Jurkat (AUPR, 0.73; AUC, 0.71) cells, while no existing methods exceed 0.62 AUPR and AUC. The prediction performance of MLEC-iSL is validated in a CDKO experiment in 22Rv1 cells, yielding a 46.8% SL rate among 987 selected gene pairs. The screen also reveals SL dependency between apoptosis and mitosis cell death pathways.
Collapse
Affiliation(s)
- Kunjie Fan
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, United States
| | - Birkan Gökbağ
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, United States
| | - Shan Tang
- Department of Biomedical Informatics, College of Pharmacy, The Ohio State University, 500 W. 12 ave, Columbus, OH 43210, United States
| | - Shangjia Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, United States
| | - Yirui Huang
- Department of Biomedical Informatics, College of Pharmacy, The Ohio State University, 500 W. 12 ave, Columbus, OH 43210, United States
| | - Lingling Wang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, United States
| | - Lijun Cheng
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, United States
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, United States
- Department of Biomedical Informatics, College of Pharmacy, The Ohio State University, 500 W. 12 ave, Columbus, OH 43210, United States
| |
Collapse
|
4
|
Liao S, Jia S, Yue Y, Zeng H, Lin J, Liu P. Advancements in pH-Responsive nanoparticles for osteoarthritis treatment: Opportunities and challenges. Front Bioeng Biotechnol 2024; 12:1426794. [PMID: 39036562 PMCID: PMC11260422 DOI: 10.3389/fbioe.2024.1426794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/06/2024] [Indexed: 07/23/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative disease linked to aging and obesity. The global aging population has led to an increasing number of OA patients, imposing a significant economic burden on society. Traditional drugs treatment methods often fail to achieve satisfactory outcomes. With the rapid advancement of nanomaterial delivery systems, numerous studies have focused on utilizing nanomaterials as carriers to achieve efficient OA treatment by effectively loading and delivering bioactive ingredients (e.g., drugs, nucleic acids) tailored to the unique pathological conditions, such as the weakly acidic microenvironment of synovial fluid in OA patients. This review highlights the latest advancements in the use of pH-responsive nanoparticles for OA treatment, emphasizing the principle of targeted drug delivery leveraging the acidic microenvironment of inflamed joints. It further discusses the composition, synthesis, response mechanism, target selection, application, and recent research findings of nanoparticles, while also addressing the challenges and future directions in this promising field.
Collapse
Affiliation(s)
- Shuai Liao
- Department of Bone and Joint Surgery, Peking
University Shenzhen Hospital, Shenzhen,
China
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen University School of Medicine,
Shenzhen, China
| | - Shicheng Jia
- Department of Sport Medicine, Peking
University Shenzhen Hospital, Shenzhen,
China
| | - Yaohang Yue
- Department of Bone and Joint Surgery, Peking
University Shenzhen Hospital, Shenzhen,
China
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials
Research, Shenzhen,
China
| | - Hui Zeng
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials
Research, Shenzhen,
China
| | - Jianjin Lin
- Department of Sport Medicine, Peking
University Shenzhen Hospital, Shenzhen,
China
| | - Peng Liu
- Department of Bone and Joint Surgery, Peking
University Shenzhen Hospital, Shenzhen,
China
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials
Research, Shenzhen,
China
| |
Collapse
|
5
|
Kim Y, Han Y, Hopper C, Lee J, Joo JI, Gong JR, Lee CK, Jang SH, Kang J, Kim T, Cho KH. A gray box framework that optimizes a white box logical model using a black box optimizer for simulating cellular responses to perturbations. CELL REPORTS METHODS 2024; 4:100773. [PMID: 38744288 PMCID: PMC11133856 DOI: 10.1016/j.crmeth.2024.100773] [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: 05/13/2023] [Revised: 03/19/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024]
Abstract
Predicting cellular responses to perturbations requires interpretable insights into molecular regulatory dynamics to perform reliable cell fate control, despite the confounding non-linearity of the underlying interactions. There is a growing interest in developing machine learning-based perturbation response prediction models to handle the non-linearity of perturbation data, but their interpretation in terms of molecular regulatory dynamics remains a challenge. Alternatively, for meaningful biological interpretation, logical network models such as Boolean networks are widely used in systems biology to represent intracellular molecular regulation. However, determining the appropriate regulatory logic of large-scale networks remains an obstacle due to the high-dimensional and discontinuous search space. To tackle these challenges, we present a scalable derivative-free optimizer trained by meta-reinforcement learning for Boolean network models. The logical network model optimized by the trained optimizer successfully predicts anti-cancer drug responses of cancer cell lines, while simultaneously providing insight into their underlying molecular regulatory mechanisms.
Collapse
Affiliation(s)
- Yunseong Kim
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Younghyun Han
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Corbin Hopper
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jonghoon Lee
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jae Il Joo
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jeong-Ryeol Gong
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Chun-Kyung Lee
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Seong-Hoon Jang
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Junsoo Kang
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Taeyoung Kim
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Kwang-Hyun Cho
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea.
| |
Collapse
|
6
|
Lee J, Kim N, Cho KH. Decoding the principle of cell-fate determination for its reverse control. NPJ Syst Biol Appl 2024; 10:47. [PMID: 38710700 PMCID: PMC11074314 DOI: 10.1038/s41540-024-00372-2] [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: 12/11/2023] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Understanding and manipulating cell fate determination is pivotal in biology. Cell fate is determined by intricate and nonlinear interactions among molecules, making mathematical model-based quantitative analysis indispensable for its elucidation. Nevertheless, obtaining the essential dynamic experimental data for model development has been a significant obstacle. However, recent advancements in large-scale omics data technology are providing the necessary foundation for developing such models. Based on accumulated experimental evidence, we can postulate that cell fate is governed by a limited number of core regulatory circuits. Following this concept, we present a conceptual control framework that leverages single-cell RNA-seq data for dynamic molecular regulatory network modeling, aiming to identify and manipulate core regulatory circuits and their master regulators to drive desired cellular state transitions. We illustrate the proposed framework by applying it to the reversion of lung cancer cell states, although it is more broadly applicable to understanding and controlling a wide range of cell-fate determination processes.
Collapse
Affiliation(s)
- Jonghoon Lee
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Namhee Kim
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- biorevert, Inc., Daejeon, Republic of Korea
| | - Kwang-Hyun Cho
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
7
|
Li H, Liu B. BioSeq-Diabolo: Biological sequence similarity analysis using Diabolo. PLoS Comput Biol 2023; 19:e1011214. [PMID: 37339155 DOI: 10.1371/journal.pcbi.1011214] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 05/24/2023] [Indexed: 06/22/2023] Open
Abstract
As the key for biological sequence structure and function prediction, disease diagnosis and treatment, biological sequence similarity analysis has attracted more and more attentions. However, the exiting computational methods failed to accurately analyse the biological sequence similarities because of the various data types (DNA, RNA, protein, disease, etc) and their low sequence similarities (remote homology). Therefore, new concepts and techniques are desired to solve this challenging problem. Biological sequences (DNA, RNA and protein sequences) can be considered as the sentences of "the book of life", and their similarities can be considered as the biological language semantics (BLS). In this study, we are seeking the semantics analysis techniques derived from the natural language processing (NLP) to comprehensively and accurately analyse the biological sequence similarities. 27 semantics analysis methods derived from NLP were introduced to analyse biological sequence similarities, bringing new concepts and techniques to biological sequence similarity analysis. Experimental results show that these semantics analysis methods are able to facilitate the development of protein remote homology detection, circRNA-disease associations identification and protein function annotation, achieving better performance than the other state-of-the-art predictors in the related fields. Based on these semantics analysis methods, a platform called BioSeq-Diabolo has been constructed, which is named after a popular traditional sport in China. The users only need to input the embeddings of the biological sequence data. BioSeq-Diabolo will intelligently identify the task, and then accurately analyse the biological sequence similarities based on biological language semantics. BioSeq-Diabolo will integrate different biological sequence similarities in a supervised manner by using Learning to Rank (LTR), and the performance of the constructed methods will be evaluated and analysed so as to recommend the best methods for the users. The web server and stand-alone package of BioSeq-Diabolo can be accessed at http://bliulab.net/BioSeq-Diabolo/server/.
Collapse
Affiliation(s)
- Hongliang Li
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing, China
| | - Bin Liu
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
8
|
Zhang M, Peng X, Ding Y, Ke X, Ren K, Xin Q, Qin M, Xie J, Li J. A cyclic brush zwitterionic polymer based pH-responsive nanocarrier-mediated dual drug delivery system with lubrication maintenance for osteoarthritis treatment. MATERIALS HORIZONS 2023. [PMID: 37078123 DOI: 10.1039/d3mh00218g] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Enhanced joint synergistic lubrication combined with anti-inflammatory therapy is an effective strategy to delay the progression of early osteoarthritis (OA) but has been rarely reported. The hydration lubrication of zwitterions and inherent super-lubrication properties of the cyclic brush, as well as the enhancement of the steric stability of the cyclic topology, can effectively improve the drug loading and utilization; herein we report a pH-responsive cyclic brush zwitterionic polymer (CB) with SBMA and DMAEMA as brushes and a cyclic polymer (c-P(HEMA)) as the core template, possessing a low coefficient of friction (0.017). After loading with hydrophobic curcumin and hydrophilic loxoprofen sodium it demonstrates high drug-loading efficiency. In vitro and in vivo experiments confirmed the triple function of the CB on superlubrication, sequence controlled release and anti-inflammatory effects demonstrated by Micro CT, histological analysis and qRT-PCR. Overall, the CB is a promising long-acting lubricating therapeutic agent, with potential for OA treatment or other diseases.
Collapse
Affiliation(s)
- Miao Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Xu Peng
- Experimental and Research Animal Institute, Sichuan University, Chengdu 610065, P. R. China
| | - Yuan Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Xiang Ke
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Kai Ren
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Qiangwei Xin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Meng Qin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Jing Xie
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
- Med-X Center for Materials, Sichuan University, Chengdu 610041, P. R. China
| |
Collapse
|
9
|
Wang Y, Liu C, Qiao X, Han X, Liu ZP. PKI: A bioinformatics method of quantifying the importance of nodes in gene regulatory network via a pseudo knockout index. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194911. [PMID: 36804477 DOI: 10.1016/j.bbagrm.2023.194911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/09/2023] [Accepted: 01/30/2023] [Indexed: 02/18/2023]
Abstract
BACKGROUND Gene regulatory network (GRN) is a model that characterizes the complex relationships between genes and thereby provides an informatics environment to measure the importance of nodes. The evaluation of important nodes in a GRN can effectively refer to their functional implications severing as key players in particular biological processes, such as master regulator and driver gene. Currently, it is mainly based on network topological parameters and focuses only on evaluating a single node individually. However, genes and products play their functions by interacting with each other. It is worth noting that the effects of gene combinations in GRN are not simply additive. Key combinations discovery is of significance in revealing gene sets with important functions. Recently, with the development of single-cell RNA-sequencing (scRNA-seq) technology, we can quantify gene expression profiles of individual cells that provide the potential to identify crucial nodes in gene regulations regarding specific condition, e.g., stem cell differentiation. RESULTS In this paper, we propose a bioinformatics method, called Pseudo Knockout Importance (PKI), to quantify the importance of node and node sets in a specific GRN structure using time-course scRNA-seq data. First, we construct ordinary differential equations to approach the gene regulations during cell differentiation. Then we design gene pseudo knockout experiments and define PKI score evaluation criteria based on the coefficient of determination. The importance of nodes can be described as the influence on the ODE system of removing variables. For key gene combinations, PKI is derived as a combinatorial optimization problem of quantifying the in silico gene knockout effects. CONCLUSIONS Here, we focus our analyses on the specific GRN of embryonic stem cells with time series gene expression profile. To verify the effectiveness and advantage of PKI method, we compare its node importance rankings with other twelve kinds of centrality-based methods, such as degree and Latora closeness. For key node combinations, we compare the results with the method based on minimum dominant set. Moreover, the famous combinations of transcription factors in induced pluripotent stem cell are also employed to verify the vital gene combinations identified by PKI. These results demonstrate the reliability and superiority of the proposed method.
Collapse
Affiliation(s)
- Yijuan Wang
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Chao Liu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xu Qiao
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Xianhua Han
- Faculty of Science, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Zhi-Ping Liu
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China.
| |
Collapse
|
10
|
Cheng X, Amanullah M, Liu W, Liu Y, Pan X, Zhang H, Xu H, Liu P, Lu Y. WMDS.net: a network control framework for identifying key players in transcriptome programs. Bioinformatics 2023; 39:7023921. [PMID: 36727489 PMCID: PMC9925106 DOI: 10.1093/bioinformatics/btad071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 01/16/2023] [Accepted: 02/01/2023] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Mammalian cells can be transcriptionally reprogramed to other cellular phenotypes. Controllability of such complex transitions in transcriptional networks underlying cellular phenotypes is an inherent biological characteristic. This network controllability can be interpreted by operating a few key regulators to guide the transcriptional program from one state to another. Finding the key regulators in the transcriptional program can provide key insights into the network state transition underlying cellular phenotypes. RESULTS To address this challenge, here, we proposed to identify the key regulators in the transcriptional co-expression network as a minimum dominating set (MDS) of driver nodes that can fully control the network state transition. Based on the theory of structural controllability, we developed a weighted MDS network model (WMDS.net) to find the driver nodes of differential gene co-expression networks. The weight of WMDS.net integrates the degree of nodes in the network and the significance of gene co-expression difference between two physiological states into the measurement of node controllability of the transcriptional network. To confirm its validity, we applied WMDS.net to the discovery of cancer driver genes in RNA-seq datasets from The Cancer Genome Atlas. WMDS.net is powerful among various cancer datasets and outperformed the other top-tier tools with a better balance between precision and recall. AVAILABILITY AND IMPLEMENTATION https://github.com/chaofen123/WMDS.net. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Xiang Cheng
- Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310006, China.,Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China
| | - Md Amanullah
- Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China.,Department of Respiratory Medicine, Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Weigang Liu
- Department of Respiratory Medicine, Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yi Liu
- Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China.,Department of Respiratory Medicine, Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Xiaoqing Pan
- Department of Mathematics, Shanghai Normal University, Xuhui 200234, China
| | - Honghe Zhang
- Department of Pathology, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Haiming Xu
- Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China
| | - Pengyuan Liu
- Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310006, China.,Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Cancer Center, Zhejiang University, Hangzhou 310029, China
| | - Yan Lu
- Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310006, China.,Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China.,Cancer Center, Zhejiang University, Hangzhou 310029, China
| |
Collapse
|
11
|
Pan X, Yun J, Coban Akdemir ZH, Jiang X, Wu E, Huang JH, Sahni N, Yi SS. AI-DrugNet: A network-based deep learning model for drug repurposing and combination therapy in neurological disorders. Comput Struct Biotechnol J 2023; 21:1533-1542. [PMID: 36879885 PMCID: PMC9984442 DOI: 10.1016/j.csbj.2023.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Discovering effective therapies is difficult for neurological and developmental disorders in that disease progression is often associated with a complex and interactive mechanism. Over the past few decades, few drugs have been identified for treating Alzheimer's disease (AD), especially for impacting the causes of cell death in AD. Although drug repurposing is gaining more success in developing therapeutic efficacy for complex diseases such as common cancer, the complications behind AD require further study. Here, we developed a novel prediction framework based on deep learning to identify potential repurposed drug therapies for AD, and more importantly, our framework is broadly applicable and may generalize to identifying potential drug combinations in other diseases. Our prediction framework is as follows: we first built a drug-target pair (DTP) network based on multiple drug features and target features, as well as the associations between DTP nodes where drug-target pairs are the DTP nodes and the associations between DTP nodes are represented as the edges in the AD disease network; furthermore, we incorporated the drug-target feature from the DTP network and the relationship information between drug-drug, target-target, drug-target within and outside of drug-target pairs, representing each drug-combination as a quartet to generate corresponding integrated features; finally, we developed an AI-based Drug discovery Network (AI-DrugNet), which exhibits robust predictive performance. The implementation of our network model help identify potential repurposed and combination drug options that may serve to treat AD and other diseases.
Collapse
Affiliation(s)
- Xingxin Pan
- Livestrong Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jun Yun
- Oden Institute for Computational Engineering and Sciences (ICES), The University of Texas at Austin, Austin, TX 78712, USA
| | - Zeynep H. Coban Akdemir
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaoqian Jiang
- School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Erxi Wu
- Livestrong Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
- Neuroscience Institute and Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA
- Department of Surgery, Texas A & M University Health Science Center, College of Medicine, Temple, TX 76508, USA
- Department of Pharmaceutical Sciences, Texas A & M University Health Science Center, College of Pharmacy, College Station, TX 77843, USA
| | - Jason H. Huang
- Neuroscience Institute and Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA
- Department of Surgery, Texas A & M University Health Science Center, College of Medicine, Temple, TX 76508, USA
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Quantitative and Computational Biosciences Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - S. Stephen Yi
- Livestrong Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
- Oden Institute for Computational Engineering and Sciences (ICES), The University of Texas at Austin, Austin, TX 78712, USA
- Interdisciplinary Life Sciences Graduate Programs (ILSGP), College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
12
|
Fan K, Tang S, Gökbağ B, Cheng L, Li L. Multi-view graph convolutional network for cancer cell-specific synthetic lethality prediction. Front Genet 2023; 13:1103092. [PMID: 36699450 PMCID: PMC9868610 DOI: 10.3389/fgene.2022.1103092] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
Synthetic lethal (SL) genetic interactions have been regarded as a promising focus for investigating potential targeted therapeutics to tackle cancer. However, the costly investment of time and labor associated with wet-lab experimental screenings to discover potential SL relationships motivates the development of computational methods. Although graph neural network (GNN) models have performed well in the prediction of SL gene pairs, existing GNN-based models are not designed for predicting cancer cell-specific SL interactions that are more relevant to experimental validation in vitro. Besides, neither have existing methods fully utilized diverse graph representations of biological features to improve prediction performance. In this work, we propose MVGCN-iSL, a novel multi-view graph convolutional network (GCN) model to predict cancer cell-specific SL gene pairs, by incorporating five biological graph features and multi-omics data. Max pooling operation is applied to integrate five graph-specific representations obtained from GCN models. Afterwards, a deep neural network (DNN) model serves as the prediction module to predict the SL interactions in individual cancer cells (iSL). Extensive experiments have validated the model's successful integration of the multiple graph features and state-of-the-art performance in the prediction of potential SL gene pairs as well as generalization ability to novel genes.
Collapse
Affiliation(s)
- Kunjie Fan
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Shan Tang
- College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Birkan Gökbağ
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Lijun Cheng
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Lang Li
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States,College of Pharmacy, The Ohio State University, Columbus, OH, United States,*Correspondence: Lang Li,
| |
Collapse
|
13
|
Wang B, Liu Q, Li Y, Chen L, Guan S, Li R, Li B, Yu Y, Liu J, Zhang Y, Wang Z. Optimizing Genomic Control in Hit Network-Target Set Model Associations with Lung Adenocarcinoma. J Cancer 2023; 14:129-139. [PMID: 36605489 PMCID: PMC9809338 DOI: 10.7150/jca.78138] [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: 08/18/2022] [Accepted: 11/07/2022] [Indexed: 01/04/2023] Open
Abstract
Background: Hit network-target sets (HNSs), compiled sets of different network nodes of the same type, are available and play a significant role in cancer development but are notoriously more difficult to select than a single target. This is due to a combination of challenges attributed to the differential of node interactions, node heterogeneity, and the limitations of node-hit information. Methods: In this study, we constructed a lung adenocarcinoma regulatory network using TCGA data and obtained different HNSs of driver nodes (DNs), core modules (CMs) and core nodes (CNs) through three kinds of methods. Then, the optimized HNS (OHNS) was obtained by integrating CMs, CNs and DNs, and the performance of different HNSs was evaluated according to network structure importance, control capability, and clinical value. Results: We found that the OHNS has two main advantages, the central location of the network and the ability to control the network, and it plays an important role in the disease network through its multifaceted capabilities. Three unique pathways were discovered in the OHNS, which is consistent with previous experiments. Additionally, 13 genes were predicted to play roles in risk prognosis, disease drivers, and cell perturbation effects of lung adenocarcinoma, of which 12 may be candidates for new drugs and biomarkers of lung adenocarcinoma. Conclusion: This study can help us understand and control a network more effectively to determine the development trend of a disease, design effective multitarget drugs, and guide the therapeutic community to optimize appropriate strategies according to different research aims in cancer treatment.
Collapse
Affiliation(s)
- Bo Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China
| | - Qiong Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China.,Postdoctoral Research Station, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yanda Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China.,The Open University of China 75 Fuxing Rd, Haidian District, Beijing 100039
| | - Lin Chen
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China.,The Second Affiliated Hospital of Zhejiang Chinese Medical University (ZhejiangXinhuaHospital), Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shuang Guan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China
| | - Rong Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China.,Rongcheng Hospital of Traditional Chinese Medicine, Weihai 264399, China
| | - Bing Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China.,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China
| | - Yanan Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China
| | - Yingying Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang, Beijing 100700, China.,✉ Corresponding authors: Yingying Zhang and Zhong Wang contributed equally to this work. Associate Professor Yingying Zhang, National Clinical Study Institution for Drugs, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongzhimen, Beijing 100700, China. Tel: 86-10-84017330; Fax: 86-10-84013115; E-mail: . Professor Zhong Wang, Pharmacological Research Laboratory, Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No. 16, Dongzhimen Nanxiaojie, Beijing 100700, China. Tel: 86-10-64014411-3308; Fax: 86-10-84032881; E-mail:
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China.,✉ Corresponding authors: Yingying Zhang and Zhong Wang contributed equally to this work. Associate Professor Yingying Zhang, National Clinical Study Institution for Drugs, Dongzhimen Hospital, Beijing University of Chinese Medicine, Dongzhimen, Beijing 100700, China. Tel: 86-10-84017330; Fax: 86-10-84013115; E-mail: . Professor Zhong Wang, Pharmacological Research Laboratory, Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No. 16, Dongzhimen Nanxiaojie, Beijing 100700, China. Tel: 86-10-64014411-3308; Fax: 86-10-84032881; E-mail:
| |
Collapse
|
14
|
Liu C, Liu C, Bai Y, Wang J, Tian W. Drug Self-Delivery Systems: Molecule Design, Construction Strategy, and Biological Application. Adv Healthc Mater 2022; 12:e2202769. [PMID: 36538727 DOI: 10.1002/adhm.202202769] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/29/2022] [Indexed: 02/01/2023]
Abstract
Drug self-delivery systems (DSDSs) offer new ways to create novel drug delivery systems (DDSs). In typical DSDSs, therapeutic reagents are not considered passive cargos but active delivery agents of actionable targets. As an advanced drug delivery strategy, DSDSs with positive cooperativity of both free drugs and nanocarriers exhibit the clear merits of unprecedented drug-loading capacity, minimized systemic toxicity, and flexible preparation of nanoscale deliverables for passive targeted therapy. This review highlights the recent advances and future trends in DSDSs on the basis of two differently constructed structures: covalent and noncovalent bond-based DSDSs. Specifically, various chemical and architectural designs, fabrication strategies, and responsive and functional features are comprehensively discussed for these two types of DSDSs. In addition, additional comments on the current development status of DSDSs and the potential applications of their molecular designs are presented in the corresponding discussion. Finally, the promising potential of DSDSs in biological applications is revealed and the relationship between preliminary molecular design of DSDSs and therapeutic effects of subsequent DSDSs biological applications is clarified.
Collapse
Affiliation(s)
- Chengfei Liu
- Shaanxi Key Laboratory of Macromolecular Science and Technology, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Caiping Liu
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, China
| | - Yang Bai
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, China
| | - Jingxia Wang
- Shaanxi Key Laboratory of Macromolecular Science and Technology, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Wei Tian
- Shaanxi Key Laboratory of Macromolecular Science and Technology, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| |
Collapse
|
15
|
Esmaeilzadeh AA, Kashian M, Salman HM, Alsaffar MF, Jaber MM, Soltani S, Amiri Manjili D, Ilhan A, Bahrami A, Kastelic JW. Identify Biomarkers and Design Effective Multi-Target Drugs in Ovarian Cancer: Hit Network-Target Sets Model Optimizing. BIOLOGY 2022; 11:1851. [PMID: 36552360 PMCID: PMC9776135 DOI: 10.3390/biology11121851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Epithelial ovarian cancer (EOC) is highly aggressive with poor patient outcomes, and a deeper understanding of ovarian cancer tumorigenesis could help guide future treatment development. We proposed an optimized hit network-target sets model to systematically characterize the underlying pathological mechanisms and intra-tumoral heterogeneity in human ovarian cancer. Using TCGA data, we constructed an epithelial ovarian cancer regulatory network in this study. We use three distinct methods to produce different HNSs for identification of the driver genes/nodes, core modules, and core genes/nodes. Following the creation of the optimized HNS (OHNS) by the integration of DN (driver nodes), CM (core module), and CN (core nodes), the effectiveness of various HNSs was assessed based on the significance of the network topology, control potential, and clinical value. Immunohistochemical (IHC), qRT-PCR, and Western blotting were adopted to measure the expression of hub genes and proteins involved in epithelial ovarian cancer (EOC). We discovered that the OHNS has two key advantages: the network's central location and controllability. It also plays a significant role in the illness network due to its wide range of capabilities. The OHNS and clinical samples revealed the endometrial cancer signaling, and the PI3K/AKT, NER, and BMP pathways. MUC16, FOXA1, FBXL2, ARID1A, COX15, COX17, SCO1, SCO2, NDUFA4L2, NDUFA, and PTEN hub genes were predicted and may serve as potential candidates for new treatments and biomarkers for EOC. This research can aid in better capturing the disease progression, the creation of potent multi-target medications, and the direction of the therapeutic community in the optimization of effective treatment regimens by various research objectives in cancer treatment.
Collapse
Affiliation(s)
| | - Mahdis Kashian
- Department of Obstetrics and Gynecology, Medical College of Iran University, Tehran 14535, Iran;
| | - Hayder Mahmood Salman
- Department of Computer Science, Al-Turath University College Al Mansour, Baghdad 10011, Iraq;
| | - Marwa Fadhil Alsaffar
- Medical Laboratory Techniques Department, AL-Mustaqbal University College, Hillah 51001, Iraq;
| | - Mustafa Musa Jaber
- Computer Techniques Engineering Department, Dijlah University College, Baghdad 00964, Iraq;
- Computer Techniques Engineering Department, Al-Farahidi University, Baghdad 10011, Iraq
| | - Siamak Soltani
- Department of Forensic Medicine, School of Medicine, Iran University of Medical Sciences, Tehran 14535, Iran;
| | - Danial Amiri Manjili
- Department of Infectious Disease, School of Medicine, Babol University of Medical Sciences, Babol 47414, Iran
| | - Ahmet Ilhan
- Department of Medical Biochemistry, Faculty of Medicine, Cukurova University, Adana 01330, Turkey
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj 1417643184, Iran;
- Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, 80333 Munich, Germany
| | - John W. Kastelic
- Department of Health, University of Calgary, Calgary, AB T2N 1N4, Canada;
| |
Collapse
|
16
|
Tang S, Gökbağ B, Fan K, Shao S, Huo Y, Wu X, Cheng L, Li L. Synthetic lethal gene pairs: Experimental approaches and predictive models. Front Genet 2022; 13:961611. [PMID: 36531238 PMCID: PMC9751344 DOI: 10.3389/fgene.2022.961611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/07/2022] [Indexed: 03/27/2024] Open
Abstract
Synthetic lethality (SL) refers to a genetic interaction in which the simultaneous perturbation of two genes leads to cell or organism death, whereas viability is maintained when only one of the pair is altered. The experimental exploration of these pairs and predictive modeling in computational biology contribute to our understanding of cancer biology and the development of cancer therapies. We extensively reviewed experimental technologies, public data sources, and predictive models in the study of synthetic lethal gene pairs and herein detail biological assumptions, experimental data, statistical models, and computational schemes of various predictive models, speculate regarding their influence on individual sample- and population-based synthetic lethal interactions, discuss the pros and cons of existing SL data and models, and highlight potential research directions in SL discovery.
Collapse
Affiliation(s)
- Shan Tang
- College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Birkan Gökbağ
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Kunjie Fan
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Shuai Shao
- College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Yang Huo
- Indiana University, Bloomington, IN, United States
| | - Xue Wu
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Lijun Cheng
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
17
|
Zhong J, Zhou W, Kang J, Fang Z, Xie M, Xiao Q, Peng W. DNRLCNN: A CNN Framework for Identifying MiRNA-Disease Associations Using Latent Feature Matrix Extraction with Positive Samples. Interdiscip Sci 2022; 14:607-622. [PMID: 35428965 DOI: 10.1007/s12539-022-00509-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 06/14/2023]
Abstract
Emerging evidence indicates that miRNAs have strong relationships with many human diseases. Investigating the associations will contribute to elucidating the activities of miRNAs and pathogenesis mechanisms, and providing new opportunities for disease diagnosis and drug discovery. Therefore, it is of significance to identify potential associations between miRNAs and diseases. The existing databases about the miRNA-disease associations (MDAs) only provide the known MDAs, which can be regarded as positive samples. However, the unknown MDAs are not sufficient to regard as reliable negative samples. To deal with this uncertainty, we proposed a convolutional neural network (CNN) framework, named DNRLCNN, based on a latent feature matrix extracted by only positive samples to predict MDAs. First, by only considering the positive samples into the calculation process, we captured the latent feature matrix for complex interactions between miRNAs and diseases in low-dimensional space. Then, we constructed a feature vector for each miRNA and disease pair based on the feature representation. Finally, we adopted a modified CNN for the feature vector to predict MDAs. As a result, our model achieves better performance than other state-of-the-art methods which based CNN in fivefold cross-validation on both miRNA-disease association prediction task (average AUC of 0.9030) and miRNA-phenotype association prediction task (average AUC of 0. 9442). In addition, we carried out case studies on two human diseases, and all the top-50 predicted miRNAs for lung neoplasms are confirmed by HMDD v3.2 and dbDEMC 2.0 databases, 98% of the top-50 predicted miRNAs for heart failure are confirmed. The experiment results show that our model has the capability of inferring potential disease-related miRNAs.
Collapse
Affiliation(s)
- Jiancheng Zhong
- College of Information Science and Engineering, Hunan Normal University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, 410083, China
| | - Wubin Zhou
- College of Information Science and Engineering, Hunan Normal University, Changsha, 410083, China
| | - Jiedong Kang
- College of Information Science and Engineering, Hunan Normal University, Changsha, 410083, China
| | - Zhuo Fang
- College of Information Science and Engineering, Hunan Normal University, Changsha, 410083, China
| | - Minzhu Xie
- College of Information Science and Engineering, Hunan Normal University, Changsha, 410083, China
| | - Qiu Xiao
- College of Information Science and Engineering, Hunan Normal University, Changsha, 410083, China.
| | - Wei Peng
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
18
|
Wang J, Wu M, Huang X, Wang L, Zhang S, Liu H, Zheng J. SynLethDB 2.0: a web-based knowledge graph database on synthetic lethality for novel anticancer drug discovery. Database (Oxford) 2022; 2022:6585691. [PMID: 35562840 PMCID: PMC9216587 DOI: 10.1093/database/baac030] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/04/2022] [Accepted: 04/24/2022] [Indexed: 11/30/2022]
Abstract
Two genes are synthetic lethal if mutations in both genes result in impaired cell viability, while mutation of either gene does not affect the cell survival. The potential usage of synthetic lethality (SL) in anticancer therapeutics has attracted many researchers to identify synthetic lethal gene pairs. To include newly identified SLs and more related knowledge, we present a new version of the SynLethDB database to facilitate the discovery of clinically relevant SLs. We extended the first version of SynLethDB database significantly by including new SLs identified through Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) screening, a knowledge graph about human SLs, a new web interface, etc. Over 16 000 new SLs and 26 types of other relationships have been added, encompassing relationships among 14 100 genes, 53 cancers, 1898 drugs, etc. Moreover, a brand-new web interface has been developed to include modules such as SL query by disease or compound, SL partner gene set enrichment analysis and knowledge graph browsing through a dynamic graph viewer. The data can be downloaded directly from the website or through the RESTful Application Programming Interfaces (APIs). Database URL: https://synlethdb.sist.shanghaitech.edu.cn/v2.
Collapse
Affiliation(s)
- Jie Wang
- School of Information Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Min Wu
- Institute for Infocomm Research, Agency for Science, Technology and Research (A*STAR), 1 Fusionopolis Way, Singapore 138632, Singapore
| | - Xuhui Huang
- School of Computing, National University of Singapore, Computing 1, 13 Computing Drive, Singapore 117417, Singapore
| | - Li Wang
- School of Information Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Sophia Zhang
- College of Agriculture and Life Sciences, Cornell University, 260 Roberts Hall, Ithaca, NY 14853, USA
| | - Hui Liu
- School of Computer Science and Technology, Nanjing Tech University, 30 Puzhu Road, Nanjing 211816, China
| | - Jie Zheng
- School of Information Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China.,Shanghai Engineering Research Center of Intelligent Vision and Imaging, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| |
Collapse
|
19
|
Construction of disease-specific cytokine profiles by associating disease genes with immune responses. PLoS Comput Biol 2022; 18:e1009497. [PMID: 35404985 PMCID: PMC9022887 DOI: 10.1371/journal.pcbi.1009497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 04/21/2022] [Accepted: 03/17/2022] [Indexed: 12/29/2022] Open
Abstract
The pathogenesis of many inflammatory diseases is a coordinated process involving metabolic dysfunctions and immune response—usually modulated by the production of cytokines and associated inflammatory molecules. In this work, we seek to understand how genes involved in pathogenesis which are often not associated with the immune system in an obvious way communicate with the immune system. We have embedded a network of human protein-protein interactions (PPI) from the STRING database with 14,707 human genes using feature learning that captures high confidence edges. We have found that our predicted Association Scores derived from the features extracted from STRING’s high confidence edges are useful for predicting novel connections between genes, thus enabling the construction of a full map of predicted associations for all possible pairs between 14,707 human genes. In particular, we analyzed the pattern of associations for 126 cytokines and found that the six patterns of cytokine interaction with human genes are consistent with their functional classifications. To define the disease-specific roles of cytokines we have collected gene sets for 11,944 diseases from DisGeNET. We used these gene sets to predict disease-specific gene associations with cytokines by calculating the normalized average Association Scores between disease-associated gene sets and the 126 cytokines; this creates a unique profile of inflammatory genes (both known and predicted) for each disease. We validated our predicted cytokine associations by comparing them to known associations for 171 diseases. The predicted cytokine profiles correlate (p-value<0.0003) with the known ones in 95 diseases. We further characterized the profiles of each disease by calculating an “Inflammation Score” that summarizes different modes of immune responses. Finally, by analyzing subnetworks formed between disease-specific pathogenesis genes, hormones, receptors, and cytokines, we identified the key genes responsible for interactions between pathogenesis and inflammatory responses. These genes and the corresponding cytokines used by different immune disorders suggest unique targets for drug discovery. The success of anti-TNF treatment in multiple inflammatory diseases suggest that there is a shared cytokine framework that defines highly conserved mechanisms of inflammation. However, clinical trials testing the efficacy of new cytokine inhibitors suggest a more complex set of interacting cytokine mechanisms that are associated with different diseases. In this work, we aim to define the disease-specific role of cytokines that mediate pathogenesis and inflammatory processes, focusing on autoimmune diseases. We hypothesize that specific clinical phenotypes result from the interactions between disease-specific cytokines and disease-related genes (identified through genetics, transcriptomics, and analysis of metabolic dysfunctions), even though they also may share a common cytokine elements and conserved mechanisms of inflammation. We have developed novel network methods that show a robust ability to identify differential associations between characteristic cytokines and genetics factors contributing to pathogenesis. We have validated our methods on 171 well-studied diseases; the predicted associations between cytokines and disease modules correlate with the published data. Our predictions provide the underlying difference of molecular mechanisms that may be responsible for clinical phenotypes.
Collapse
|
20
|
Guo WF, Zhang SW, Zeng T, Akutsu T, Chen L. Network control principles for identifying personalized driver genes in cancer. Brief Bioinform 2021; 21:1641-1662. [PMID: 31711128 DOI: 10.1093/bib/bbz089] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 02/02/2023] Open
Abstract
To understand tumor heterogeneity in cancer, personalized driver genes (PDGs) need to be identified for unraveling the genotype-phenotype associations corresponding to particular patients. However, most of the existing driver-focus methods mainly pay attention on the cohort information rather than on individual information. Recent developing computational approaches based on network control principles are opening a new way to discover driver genes in cancer, particularly at an individual level. To provide comprehensive perspectives of network control methods on this timely topic, we first considered the cancer progression as a network control problem, in which the expected PDGs are altered genes by oncogene activation signals that can change the individual molecular network from one health state to the other disease state. Then, we reviewed the network reconstruction methods on single samples and introduced novel network control methods on single-sample networks to identify PDGs in cancer. Particularly, we gave a performance assessment of the network structure control-based PDGs identification methods on multiple cancer datasets from TCGA, for which the data and evaluation package also are publicly available. Finally, we discussed future directions for the application of network control methods to identify PDGs in cancer and diverse biological processes.
Collapse
Affiliation(s)
- Wei-Feng Guo
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji, 611-0011, Japan
| | - Luonan Chen
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China.,Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China.,Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| |
Collapse
|
21
|
Ding YY, Kim H, Madden K, Loftus JP, Chen GM, Allen DH, Zhang R, Xu J, Chen CH, Hu Y, Tasian SK, Tan K. Network Analysis Reveals Synergistic Genetic Dependencies for Rational Combination Therapy in Philadelphia Chromosome-Like Acute Lymphoblastic Leukemia. Clin Cancer Res 2021; 27:5109-5122. [PMID: 34210682 DOI: 10.1158/1078-0432.ccr-21-0553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/10/2021] [Accepted: 06/25/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Systems biology approaches can identify critical targets in complex cancer signaling networks to inform new therapy combinations that may overcome conventional treatment resistance. EXPERIMENTAL DESIGN We performed integrated analysis of 1,046 childhood B-ALL cases and developed a data-driven network controllability-based approach to identify synergistic key regulator targets in Philadelphia chromosome-like B-acute lymphoblastic leukemia (Ph-like B-ALL), a common high-risk leukemia subtype associated with hyperactive signal transduction and chemoresistance. RESULTS We identified 14 dysregulated network nodes in Ph-like ALL involved in aberrant JAK/STAT, Ras/MAPK, and apoptosis pathways and other critical processes. Genetic cotargeting of the synergistic key regulator pair STAT5B and BCL2-associated athanogene 1 (BAG1) significantly reduced leukemia cell viability in vitro. Pharmacologic inhibition with dual small molecule inhibitor therapy targeting this pair of key nodes further demonstrated enhanced antileukemia efficacy of combining the BCL-2 inhibitor venetoclax with the tyrosine kinase inhibitors ruxolitinib or dasatinib in vitro in human Ph-like ALL cell lines and in vivo in multiple childhood Ph-like ALL patient-derived xenograft models. Consistent with network controllability theory, co-inhibitor treatment also shifted the transcriptomic state of Ph-like ALL cells to become less like kinase-activated BCR-ABL1-rearranged (Ph+) B-ALL and more similar to prognostically favorable childhood B-ALL subtypes. CONCLUSIONS Our study represents a powerful conceptual framework for combinatorial drug discovery based on systematic interrogation of synergistic vulnerability pathways with pharmacologic inhibitor validation in preclinical human leukemia models.
Collapse
Affiliation(s)
- Yang-Yang Ding
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Hannah Kim
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, Pennsylvania
| | - Kellyn Madden
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Joseph P Loftus
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Gregory M Chen
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Hottman Allen
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ruitao Zhang
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jason Xu
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chia-Hui Chen
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Yuxuan Hu
- School of Computer Science and Technology, Xidian University, Xi'an, Shaanxi, China
| | - Sarah K Tasian
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania. .,Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kai Tan
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania. .,Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
22
|
He L, Wen S, Zhong Z, Weng S, Jiang Q, Mi H, Liu F. The Synergistic Effects of 5-Aminosalicylic Acid and Vorinostat in the Treatment of Ulcerative Colitis. Front Pharmacol 2021; 12:625543. [PMID: 34093178 PMCID: PMC8176098 DOI: 10.3389/fphar.2021.625543] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
Background: The drug 5-aminosalicylic acid (5-ASA) is the first-line therapy for the treatment of patients with mild-to-moderate ulcerative colitis (UC). However, in some cases, 5-ASA cannot achieve the desired therapeutic effects. Therefore, patients have to undergo therapies that include corticosteroids, monoclonal antibodies or immunosuppressants, which are expensive and may be accompanied by significant side effects. Synergistic drug combinations can achieve greater therapeutic effects than individual drugs while contributing to combating drug resistance and lessening toxic side effects. Thus, in this study, we sought to identify synergistic drugs that can act synergistically with 5-ASA. Methods: We started our study with protein-metabolite analysis based on peroxisome proliferator-activated receptor gamma (PPARG), the therapeutic target of 5-ASA, to identify more additional potential drug targets. Then, we further evaluated the possibility of their synergy with PPARG by integrating Kyoto Encyclopedia of Genes and Genome (KEGG) pathway enrichment analysis, pathway-pathway interaction analysis, and semantic similarity analysis. Finally, we validated the synergistic effects with in vitro and in vivo experiments. Results: The combination of 5-ASA and vorinostat (SAHA) showed lower toxicity and mRNA expression of p65 in human colonic epithelial cell lines (Caco-2 and HCT-116), and more efficiently alleviated the symptoms of dextran sulfate sodium (DSS)-induced colitis than treatment with 5-ASA and SAHA alone. Conclusion: SAHA can exert effective synergistic effects with 5-ASA in the treatment of UC. One possible mechanism of synergism may be synergistic inhibition of the nuclear factor kappa B (NF-kB) signaling pathway. Moreover, the metabolite-butyric acid may be involved.
Collapse
Affiliation(s)
- Long He
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Reserch Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuting Wen
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Reserch Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhuotai Zhong
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Senhui Weng
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qilong Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hong Mi
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengbin Liu
- Lingnan Medical Reserch Center of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Baiyun Hospital of the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
23
|
Uncovering and classifying the role of driven nodes in control of complex networks. Sci Rep 2021; 11:9627. [PMID: 33953235 PMCID: PMC8100151 DOI: 10.1038/s41598-021-88295-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 04/05/2021] [Indexed: 11/26/2022] Open
Abstract
The widely used Maximum Matching (MM) method identifies the minimum driver nodes set to control biological and technological systems. Nevertheless, it is assumed in the MM approach that one driver node can send control signal to multiple target nodes, which might not be appropriate in certain complex networks. A recent work introduced a constraint that one driver node can control one target node, and proposed a method to identify the minimum target nodes set under such a constraint. We refer such target nodes to driven nodes. However, the driven nodes may not be uniquely determined. Here, we develop a novel algorithm to classify driven nodes in control categories. Our computational analysis on a large number of biological networks indicates that the number of driven nodes is considerably larger than the number of driver nodes, not only in all examined complete plant metabolic networks but also in several key human pathways, which firstly demonstrate the importance of use of driven nodes in analysis of real-world networks.
Collapse
|
24
|
Gallage S, García-Beccaria M, Szydlowska M, Rahbari M, Mohr R, Tacke F, Heikenwalder M. The therapeutic landscape of hepatocellular carcinoma. MED 2021; 2:505-552. [PMID: 35590232 DOI: 10.1016/j.medj.2021.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/23/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023]
|
25
|
Guo WF, Zhang SW, Feng YH, Liang J, Zeng T, Chen L. Network controllability-based algorithm to target personalized driver genes for discovering combinatorial drugs of individual patients. Nucleic Acids Res 2021; 49:e37. [PMID: 33434272 PMCID: PMC8053130 DOI: 10.1093/nar/gkaa1272] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/02/2020] [Accepted: 12/22/2020] [Indexed: 12/27/2022] Open
Abstract
Multiple driver genes in individual patient samples may cause resistance to individual drugs in precision medicine. However, current computational methods have not studied how to fill the gap between personalized driver gene identification and combinatorial drug discovery for individual patients. Here, we developed a novel structural network controllability-based personalized driver genes and combinatorial drug identification algorithm (CPGD), aiming to identify combinatorial drugs for an individual patient by targeting personalized driver genes from network controllability perspective. On two benchmark disease datasets (i.e. breast cancer and lung cancer datasets), performance of CPGD is superior to that of other state-of-the-art driver gene-focus methods in terms of discovery rate among prior-known clinical efficacious combinatorial drugs. Especially on breast cancer dataset, CPGD evaluated synergistic effect of pairwise drug combinations by measuring synergistic effect of their corresponding personalized driver gene modules, which are affected by a given targeting personalized driver gene set of drugs. The results showed that CPGD performs better than existing synergistic combinatorial strategies in identifying clinical efficacious paired combinatorial drugs. Furthermore, CPGD enhanced cancer subtyping by computationally providing personalized side effect signatures for individual patients. In addition, CPGD identified 90 drug combinations candidates from SARS-COV2 dataset as potential drug repurposing candidates for recently spreading COVID-19.
Collapse
Affiliation(s)
- Wei-Feng Guo
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian 710072, China.,School of Electrical Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian 710072, China
| | - Yue-Hua Feng
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian 710072, China
| | - Jing Liang
- School of Electrical Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Tao Zeng
- CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy Science, Shanghai 200031, China
| | - Luonan Chen
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy Science, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China.,Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
26
|
Ruiz C, Zitnik M, Leskovec J. Identification of disease treatment mechanisms through the multiscale interactome. Nat Commun 2021; 12:1796. [PMID: 33741907 PMCID: PMC7979814 DOI: 10.1038/s41467-021-21770-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Most diseases disrupt multiple proteins, and drugs treat such diseases by restoring the functions of the disrupted proteins. How drugs restore these functions, however, is often unknown as a drug's therapeutic effects are not limited to the proteins that the drug directly targets. Here, we develop the multiscale interactome, a powerful approach to explain disease treatment. We integrate disease-perturbed proteins, drug targets, and biological functions into a multiscale interactome network. We then develop a random walk-based method that captures how drug effects propagate through a hierarchy of biological functions and physical protein-protein interactions. On three key pharmacological tasks, the multiscale interactome predicts drug-disease treatment, identifies proteins and biological functions related to treatment, and predicts genes that alter a treatment's efficacy and adverse reactions. Our results indicate that physical interactions between proteins alone cannot explain treatment since many drugs treat diseases by affecting the biological functions disrupted by the disease rather than directly targeting disease proteins or their regulators. We provide a general framework for explaining treatment, even when drugs seem unrelated to the diseases they are recommended for.
Collapse
Affiliation(s)
- Camilo Ruiz
- Computer Science Department, Stanford University, Stanford, CA, USA
- Bioengineering Department, Stanford University, Stanford, CA, USA
| | - Marinka Zitnik
- Biomedical Informatics Department, Harvard University, Boston, MA, USA
| | - Jure Leskovec
- Computer Science Department, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
27
|
Thng DKH, Toh TB, Chow EKH. Capitalizing on Synthetic Lethality of MYC to Treat Cancer in the Digital Age. Trends Pharmacol Sci 2021; 42:166-182. [PMID: 33422376 DOI: 10.1016/j.tips.2020.11.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023]
Abstract
Deregulation of MYC is among the most frequent oncogenic drivers of cancer. Developing targeted therapies against MYC is, therefore, one of the most critical unmet needs of cancer therapy. Unfortunately, MYC has been labelled as undruggable due to the lack of success in developing clinically relevant MYC-targeted therapies. Synthetic lethality is a promising approach that targets MYC-dependent vulnerabilities in cancer. However, translating the synthetic lethality targets to the clinics is still challenging due to the complex nature of cancers. This review highlights the most promising mechanisms of MYC synthetic lethality and how these discoveries are currently translated into the clinic. Finally, we discuss how in silico computational platforms can improve clinical success of synthetic lethality-based therapy.
Collapse
Affiliation(s)
- Dexter Kai Hao Thng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health, National University of Singapore, Singapore; The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; The N.1 Institute for Health, National University of Singapore, Singapore; The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore.
| |
Collapse
|
28
|
Chen GM, Azzam A, Ding YY, Barrett DM, Grupp SA, Tan K. Dissecting the Tumor-Immune Landscape in Chimeric Antigen Receptor T-cell Therapy: Key Challenges and Opportunities for a Systems Immunology Approach. Clin Cancer Res 2020; 26:3505-3513. [PMID: 32127393 PMCID: PMC7367708 DOI: 10.1158/1078-0432.ccr-19-3888] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/15/2020] [Accepted: 02/27/2020] [Indexed: 12/17/2022]
Abstract
The adoptive transfer of genetically engineered chimeric antigen receptor (CAR) T cells has opened a new frontier in cancer therapy. Unlike the paradigm of targeted therapies, the efficacy of CAR T-cell therapy depends not only on the choice of target but also on a complex interplay of tumor, immune, and stromal cell communication. This presents both challenges and opportunities from a discovery standpoint. Whereas cancer consortia have traditionally focused on the genomic, transcriptomic, epigenomic, and proteomic landscape of cancer cells, there is an increasing need to expand studies to analyze the interactions between tumor, immune, and stromal cell populations in their relevant anatomical and functional compartments. Here, we focus on the promising application of systems biology to address key challenges in CAR T-cell therapy, from understanding the mechanisms of therapeutic resistance in hematologic and solid tumors to addressing important clinical challenges in biomarker discovery and therapeutic toxicity. We propose a systems biology view of key clinical objectives in CAR T-cell therapy and suggest a path forward for a biomedical discovery process that leverages modern technological approaches in systems biology.
Collapse
Affiliation(s)
- Gregory M Chen
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew Azzam
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yang-Yang Ding
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - David M Barrett
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephan A Grupp
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Oncology, Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kai Tan
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Liu C, Li C, Pang C, Li M, Li H, Li P, Fan L, Liu H, Tian W. Supramolecular Drug-Drug Complex Vesicles Enable Sequential Drug Release for Enhanced Combination Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:27940-27950. [PMID: 32449351 DOI: 10.1021/acsami.0c04565] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Drug-drug self-delivery systems serving as both carriers and cargos have been explored as advanced combination chemotherapy strategies to overcome the limitations of the traditional single-drug chemotherapy. However, most known drug-drug self-delivery systems may cause a rapid increase in drug concentration when the single covalent bond is broken, thus leading to high toxicity to organs and low therapeutic efficiency against tumors. To address the above problem, in this study, a novel supramolecular drug-drug complex (SDDC) simultaneously containing both covalent and noncovalent bonds was proposed to realize the sequential release of two drugs in tumor cells for enhanced combination therapy. The SDDC could self-assemble into uniform bilayer supramolecular vesicles (SVs) with a remarkable drug loading capacity and stable drug transport. Notably, the SVs with controlled sequential release ability in tumor cells exhibited a superior synergistic effect and significantly improved therapeutic efficiency with reduced toxicity in in vivo antitumor activity and histological analyses in comparison to either individual free drugs or a mixture of two free drugs. Therefore, by combining the advantages of noncovalent interactions with the dynamic nature and stable covalent bonds, this study opens a new way for cancer therapy.
Collapse
Affiliation(s)
- Chengfei Liu
- Shaanxi Key Laboratory of Macromolecular Science and Technology, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Chunpu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Cui Pang
- Department of Pharmaceutical Chemistry and Analysis, School of Pharmacy, Air Force Medical University, 169th Changle West Road, Xi'an, 710032 Shaanxi, China
| | - Muqiong Li
- Department of Pharmaceutical Chemistry and Analysis, School of Pharmacy, Air Force Medical University, 169th Changle West Road, Xi'an, 710032 Shaanxi, China
| | - Huixin Li
- Shaanxi Key Laboratory of Macromolecular Science and Technology, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Pengxiang Li
- Shaanxi Key Laboratory of Macromolecular Science and Technology, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Li Fan
- Department of Pharmaceutical Chemistry and Analysis, School of Pharmacy, Air Force Medical University, 169th Changle West Road, Xi'an, 710032 Shaanxi, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Wei Tian
- Shaanxi Key Laboratory of Macromolecular Science and Technology, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
30
|
Guo WF, Zhang SW, Zeng T, Li Y, Gao J, Chen L. A novel network control model for identifying personalized driver genes in cancer. PLoS Comput Biol 2019; 15:e1007520. [PMID: 31765387 PMCID: PMC6901264 DOI: 10.1371/journal.pcbi.1007520] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 12/09/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Abstract
Although existing computational models have identified many common driver genes, it remains challenging to identify the personalized driver genes by using samples of an individual patient. Recently, the methods of exploiting the structure-based control principles of complex networks provide new clues for identifying minimum number of driver nodes to drive the state transition of large-scale complex networks from an initial state to the desired state. However, the structure-based network control methods cannot be directly applied to identify the personalized driver genes due to the unknown network dynamics of the personalized system. Here we proposed the personalized network control model (PNC) to identify the personalized driver genes by employing the structure-based network control principle on genetic data of individual patients. In PNC model, we firstly presented a paired single sample network construction method to construct the personalized state transition network for capturing the phenotype transitions between healthy and disease states. Then, we designed a novel structure-based network control method from the Feedback Vertex Sets-based control perspective to identify the personalized driver genes. The wide experimental results on 13 cancer datasets from The Cancer Genome Atlas firstly showed that PNC model outperforms current state-of-the-art methods, in terms of F-measures for identifying cancer driver genes enriched in the gold-standard cancer driver gene lists. Furthermore, these results showed that personalized driver genes can be explored by their network characteristics even when they are hidden factors in transcription and mutation profiles. Our PNC gives novel insights and useful tools into understanding the tumor heterogeneity in cancer. The PNC package and data resources used in this work can be freely downloaded from https://github.com/NWPU-903PR/PNC. Notably there may be unique personalized driver genes for an individual patient in cancer. Identifying personalized driver genes that lead to particular cancer initiation and progression of individual patient is one of the biggest challenges in precision medicine. However, most methods for cancer driver genes identification have focused mainly on the cohort information rather than on individual information and fail to identify personalized driver genes. We here proposed personalized network control model (PNC) to identify personalized driver genes by applying the structure based network control principle on personalized data of individual patients. By considering the progression from the healthy state to the disease state as the network control problem, our PNC aims to detect a small number of personalized driver genes that are altered in response to input signals for triggering the state transition in individual patients on expression level. The impetus behind PNC contains two main respects. One is to design a paired single sample network construction method (namely Paired-SSN) for constructing personalized state transition networks to capture the phenotypic transitions between normal and disease attractors. The other one is to develop a novel structure based network control method (namely NCUA) on personalized state transition networks for identifying personalized driver genes which can drive individual patient system state from healthy state to disease state through oncogene activations. Each part of the proposed method has been deeply examined to be efficient. Compared with other existing models, our PNC shows a higher performance in terms of F-measures of the cancer driver genes in the well-known Cancer Census Genes (CCG) and Network of Cancer Genes (NCG). The wide experimental results on multiple cancer datasets highlight that sample specific network theory and structure based network control theory can contribute to identifying personalized driver genes in cancer.
Collapse
Affiliation(s)
- Wei-Feng Guo
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian, China
- * E-mail: (S-WZ); (JG); (LC)
| | - Tao Zeng
- Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Science, Chinese Academy Science, Shanghai, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China
| | - Yan Li
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian, China
| | - Jianxi Gao
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Network Science and Technology Center, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- * E-mail: (S-WZ); (JG); (LC)
| | - Luonan Chen
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xian, China
- Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Science, Chinese Academy Science, Shanghai, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
- * E-mail: (S-WZ); (JG); (LC)
| |
Collapse
|
31
|
Liang J, Chen B, Hu J, Huang Q, Zhang D, Wan J, Hu Z, Wang B. pH and Thermal Dual-Responsive Graphene Oxide Nanocomplexes for Targeted Drug Delivery and Photothermal-Chemo/Photodynamic Synergetic Therapy. ACS APPLIED BIO MATERIALS 2019; 2:5859-5871. [DOI: 10.1021/acsabm.9b00835] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
| | | | | | | | - Dianbo Zhang
- Shandong Non-metallic Materials Institute, Jinan 250031, China
| | | | | | | |
Collapse
|