Next Article in Journal
The Sustainability Study and Exploration in the Building Commercial Complex System Based on Life Cycle Assessment (LCA)–Emergy–Carbon Emission Analysis
Previous Article in Journal
Multi-Time Scale Economic Dispatch of Integrated Electricity and Natural Gas Systems with Flexibility Constraints Based on Chance-Constrained Programming
Previous Article in Special Issue
A Study on the Mechanism of Action of Galangal in the Treatment of Gastric Cancer Using Network Pharmacology Technology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue on Network Pharmacology Modeling for Drug Discovery

1
Research Programs in Systems Oncology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
2
Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
Processes 2023, 11(7), 1988; https://doi.org/10.3390/pr11071988
Submission received: 12 June 2023 / Accepted: 27 June 2023 / Published: 30 June 2023
(This article belongs to the Special Issue Network Pharmacology Modelling for Drug Discovery)
During the process of drug discovery, many compounds have exhibited polypharmacological interactions with various biological entities [1,2,3,4]. This complexity poses a significant challenge in developing therapies to enhance efficacy with less toxicity, particularly for treating complex diseases such as cancer [5,6]. Meanwhile, the determination of bioactive compounds from natural products remains a tedious process with a low success rate, primarily due to the poor understanding of compound–activity relationships [7,8,9]. In response to this challenge, network pharmacology modeling has emerged as a promising paradigm for the next generation of drug discovery [10,11,12]. With the growing abundance of molecular data derived from both pharmacological and biological entities, the field of network pharmacology modeling has witnessed increasingly exciting applications [13,14,15,16]. This Special Issue, titled “Network Pharmacology Modelling for Drug Discovery”, which is available online at https://www.mdpi.com/journal/processes/special_issues/Network_Pharmacology_Modelling, aims to highlight the recent advances in this endeavor, with a specific focus on understanding the mechanisms of action of herb medicine. In what follows, we will provide a brief overview of the studies that were selected for the Special Issue, highlighting the database resources and computational tools that may be references for future studies.
1.
Galangal against gastric cancer
Galangal, the rhizome of the ginger plant, has been reported to relieve stomach diseases. However, its potential treatment effects on gastric cancer remain largely unexplored. In [17], the authors determined a total of 13 active compounds of galangal, as well as their potential target genes from the TCMSP database [18]. Through a protein–protein network analysis and gene ontology enrichment analysis, they found that several known gastric cancer genes indeed interact with the key targets of galangal. Furthermore, they were able to validate several ligand–receptor bindings through computational simulations, suggesting the potential of galangal in treating gastric cancer.
2.
Zhi Bai Di Huang Pill against Systemic Lupus Erythematosus (SLE)
SLE is an autoimmune disease where the body’s immune system mistakenly attacks its own organs, causing widespread inflammation and tissue damage. In [19], the authors explored the potential of a traditional Chinese medicine called Zhi Bai Di Huang Pill (ZBDHP) for the treatment of SLE. The TCMSP database was utilized to retrieve the active ingredients of ZBDHP, while their targets were predicted by the SwissTargetPrediction tool [20]. On the other hand, the SLE-associated genes were retrieved from the GeneCards [21], OMIM [22], and DisGeNET [23] databases. Further gene set enrichment analyses showed that ZBDHP may affect the PI3K, AKT, and mTOR signaling pathways. Similar databases and computational tools have also been used to study the following herb plants: Qianghuo Shengshi decoction (QHSSD) against ankylosing spondylitis [24] and Ocimum Sanctum against tuberculosis [25].
3.
Glycyrrhiza Uralensis against alcoholic liver injury
In [26], the authors explored the mechanisms of action of Glycyrrhiza uralensis, also known as Chinese liquorice, in treating alcoholic liver injury. Notably, multiple molecular docking tools were used, including AutoDock [27], PYMOL, and Discovery Studio. Using similar network pharmacology modeling approaches to those of [19], the authors provided initial evidence of Glycyrrhiza uralensis that may warrant future experimental validation using in vitro or in vivo studies.
4.
Glutinol against multiple diseases
Glutinol is a triterpenoid compound that has been reported to have a range of antidiabetic, anti-inflammatory, and anticancer effects. In [28], the authors studied the mechanisms of action as well as the ADMET properties of glutinol. Confirming first that glutinol has drug-likeness properties through using the pkCSM tool [29], the authors determined target genes from the BindingDB database [30] and their interacting proteins from the STRING database [31]. A gene ontology enrichment analysis was performed using the DAVID tool [32]. Molecular docking with MOE has further revealed top binding targets of glutinol, such as CYP19A1.
5.
Ginseng against COVID-19
Network pharmacology modeling has also been applied for treating the recent COVID-19 pandemic. In [33], the authors studied the low-molecular-weight compounds (LMWCs) from Panax Ginseng C.A. Meyer (PGCAM). Using SwissTargetPrediction and SEA [34] analyses, multiple target genes were predicted for COVID-19 and further validated using molecular docking simulations. Another antiviral study concerns quercetin against the influenza A virus (IAV), where a compound–target–pathway network has been established [35].
6.
Sochehwan against metabolic syndrome
Sochehwan is a herbal formula of traditional Korean medicine with limited knowledge on its mechanisms of action. In [36], the authors studied its effect on metabolic syndrome. Using the TCMID database [37], active compounds of Sochehwan were retrieved and screened in the STRING database for their protein targets. Notably, the authors validated the efficacy of Sochehwan in a mouse macrophage cell line, mainly through the suppression of lipopolysaccharide-induced NF-kB and MAPK inflammatory responses.
7.
Pueraria lobata against diabetes
In [38], the authors determined the active ingredients of the roots of Pueraria lobata using mass spectrometry experiments, and then retrieved their targets from TCSMP and Drugbank [39]. A related study is on obesity, where the authors used similar mass spectrometry techniques to determine the metabolites of Ilex cornuta and identified the NOD-like receptor (NLR) signaling pathway as the key target when treating obesity [40].
8.
Summary
Network pharmacology modeling is increasingly being recognized as a crucial tool for prioritizing potential drug candidates in in silico drug discovery processes, particularly for herbal medicine, which inherently contains multiple active ingredients. As we showcased the applications of network pharmacology modeling, it is also imperative to discuss their limitations. Firstly, as most of the studies have acknowledged, there is a lack of experimental validation concerning these potential drug targets. Moreover, while these studies focus on the elucidation of drug targets, they often leave unanswered how these interactions contribute to the synergistic or antagonistic effects in disease contexts. Understanding these network dynamics could significantly enhance our capacity to develop more targeted and effective treatment [41]. We anticipate that the Special Issue may bring more attention from a systems medicine perspective of drug discovery. With growing datasets about drug targets as well as their effects in multiple disease contexts, we look forward to further advances in the mechanistic modeling of network pharmacology. These developments, we believe, could improve the efficiency of drug screening and ultimately improve clinical translation as well as precision medicine [42].

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Davis, M.I.; Hunt, J.P.; Herrgard, S.; Ciceri, P.; Wodicka, L.M.; Pallares, G.; Hocker, M.; Treiber, D.K.; Zarrinkar, P.P. Comprehensive Analysis of Kinase Inhibitor Selectivity. Nat. Biotechnol. 2011, 29, 1046–1051. [Google Scholar] [CrossRef] [PubMed]
  2. Tang, J.; Szwajda, A.; Shakyawar, S.; Xu, T.; Hintsanen, P.; Wennerberg, K.; Aittokallio, T. Making Sense of Large-Scale Kinase Inhibitor Bioactivity Data Sets: A Comparative and Integrative Analysis. J. Chem. Inf. Model. 2014, 54, 735–743. [Google Scholar] [CrossRef] [PubMed]
  3. Elkins, J.M.; Fedele, V.; Szklarz, M.; Abdul Azeez, K.R.; Salah, E.; Mikolajczyk, J.; Romanov, S.; Sepetov, N.; Huang, X.-P.; Roth, B.L.; et al. Comprehensive Characterization of the Published Kinase Inhibitor Set. Nat. Biotechnol. 2016, 34, 95–103. [Google Scholar] [CrossRef] [PubMed]
  4. Lin, A.; Giuliano, C.J.; Palladino, A.; John, K.M.; Abramowicz, C.; Yuan, M.L.; Sausville, E.L.; Lukow, D.A.; Liu, L.; Chait, A.R.; et al. Off-Target Toxicity Is a Common Mechanism of Action of Cancer Drugs Undergoing Clinical Trials. Sci. Transl. Med. 2019, 11, eaaw8412. [Google Scholar] [CrossRef] [PubMed]
  5. Knight, Z.A.; Lin, H.; Shokat, K.M. Targeting the Cancer Kinome through Polypharmacology. Nat. Rev. Cancer 2010, 10, 130–137. [Google Scholar] [CrossRef] [Green Version]
  6. Antolin, A.A.; Workman, P.; Mestres, J.; Al-Lazikani, B. Polypharmacology in Precision Oncology: Current Applications and Future Prospects. Curr. Pharm. Des. 2016, 22, 6935–6945. [Google Scholar] [CrossRef] [Green Version]
  7. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C.T. Natural Products in Drug Discovery: Advances and Opportunities. Nat. Rev. Drug. Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  8. Grigalunas, M.; Brakmann, S.; Waldmann, H. Chemical Evolution of Natural Product Structure. J. Am. Chem. Soc. 2022, 144, 3314–3329. [Google Scholar] [CrossRef]
  9. Saldívar-González, F.I.; Aldas-Bulos, V.D.; Medina-Franco, J.L.; Plisson, F. Natural Product Drug Discovery in the Artificial Intelligence Era. Chemical. Sci. 2022, 13, 1526–1546. [Google Scholar] [CrossRef]
  10. Hopkins, A.L. Network Pharmacology: The next Paradigm in Drug Discovery. Nat. Chem. Biol. 2008, 4, 682–690. [Google Scholar] [CrossRef]
  11. Gu, J.; Gui, Y.; Chen, L.; Yuan, G.; Lu, H.-Z.; Xu, X. Use of Natural Products as Chemical Library for Drug Discovery and Network Pharmacology. PLoS ONE 2013, 8, e62839. [Google Scholar] [CrossRef] [Green Version]
  12. Kibble, M.; Saarinen, N.; Tang, J.; Wennerberg, K.; Mäkelä, S.; Aittokallio, T. Network Pharmacology Applications to Map the Unexplored Target Space and Therapeutic Potential of Natural Products. Nat. Prod. Rep. 2015, 32, 1249–1266. [Google Scholar] [CrossRef]
  13. Cheng, F.; Kovács, I.A.; Barabási, A.-L. Network-Based Prediction of Drug Combinations. Nat. Commun. 2019, 10, 1197. [Google Scholar] [CrossRef] [Green Version]
  14. Wang, Y.; Yang, H.; Chen, L.; Jafari, M.; Tang, J. Network-Based Modeling of Herb Combinations in Traditional Chinese Medicine. Brief. Bioinform. 2021, 22, bbab106. [Google Scholar] [CrossRef]
  15. Nogales, C.; Mamdouh, Z.M.; List, M.; Kiel, C.; Casas, A.I.; Schmidt, H.H.H.W. Network Pharmacology: Curing Causal Mechanisms Instead of Treating Symptoms. Trends Pharmacol. Sci. 2022, 43, 136–150. [Google Scholar] [CrossRef]
  16. Casas, A.I.; Hassan, A.A.; Larsen, S.J.; Gomez-Rangel, V.; Elbatreek, M.; Kleikers, P.W.M.; Guney, E.; Egea, J.; López, M.G.; Baumbach, J.; et al. From Single Drug Targets to Synergistic Network Pharmacology in Ischemic Stroke. Proc. Natl. Acad. Sci. USA 2019, 116, 7129–7136. [Google Scholar] [CrossRef] [Green Version]
  17. Tao, A.; Feng, X.; Song, Z.; Xu, R.; Zhao, Y. A Study on the Mechanism of Action of Galangal in the Treatment of Gastric Cancer Using Network Pharmacology Technology. Processes 2022, 10, 1988. [Google Scholar] [CrossRef]
  18. Ru, J.; Li, P.; Wang, J.; Zhou, W.; Li, B.; Huang, C.; Li, P.; Guo, Z.; Tao, W.; Yang, Y.; et al. TCMSP: A Database of Systems Pharmacology for Drug Discovery from Herbal Medicines. J. Cheminform. 2014, 6, 13. [Google Scholar] [CrossRef] [Green Version]
  19. Zhuang, Y.; Zhang, X.; Luo, S.; Wei, F.; Song, Y.; Lin, G.; Yao, M.; Gong, A. Exploring the Molecular Mechanism of Zhi Bai Di Huang Wan in the Treatment of Systemic Lupus Erythematosus Based on Network Pharmacology and Molecular Docking Techniques. Processes 2022, 10, 1914. [Google Scholar] [CrossRef]
  20. Daina, A.; Michielin, O.; Zoete, V. SwissTargetPrediction: Updated Data and New Features for Efficient Prediction of Protein Targets of Small Molecules. Nucleic Acids Res. 2019, 47, W357–W364. [Google Scholar] [CrossRef] [Green Version]
  21. Stelzer, G.; Rosen, N.; Plaschkes, I.; Zimmerman, S.; Twik, M.; Fishilevich, S.; Stein, T.I.; Nudel, R.; Lieder, I.; Mazor, Y.; et al. The GeneCards Suite: From Gene Data Mining to Disease Genome Sequence Analyses. Curr. Protoc. Bioinform. 2016, 54, 1.30.1–1.30.33. [Google Scholar] [CrossRef] [PubMed]
  22. Amberger, J.S.; Bocchini, C.A.; Schiettecatte, F.; Scott, A.F.; Hamosh, A. OMIM.Org: Online Mendelian Inheritance in Man (OMIM®), an Online Catalog of Human Genes and Genetic Disorders. Nucleic Acids Res. 2015, 43, D789–D798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Piñero, J.; Queralt-Rosinach, N.; Bravo, À.; Deu-Pons, J.; Bauer-Mehren, A.; Baron, M.; Sanz, F.; Furlong, L.I. DisGeNET: A Discovery Platform for the Dynamical Exploration of Human Diseases and Their Genes. Database 2015, 2015, bav028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Luo, S.; Xiao, X.; Luo, W.; Zhang, X.; Zhang, J.; Tang, S. Investigating the Molecular Mechanism of Qianghuo Shengshi Decoction in the Treatment of Ankylosing Spondylitis Based on Network Pharmacology and Molecular Docking Analysis. Processes 2022, 10, 1487. [Google Scholar] [CrossRef]
  25. Tabassum, S.; Khalid, H.R.; ul Haq, W.; Aslam, S.; Alshammari, A.; Alharbi, M.; Riaz Rajoka, M.S.; Khurshid, M.; Ashfaq, U.A. Implementation of System Pharmacology and Molecular Docking Approaches to Explore Active Compounds and Mechanism of Ocimum Sanctum against Tuberculosis. Processes 2022, 10, 298. [Google Scholar] [CrossRef]
  26. Zhu, L.; Xie, S.; Geng, Z.; Yang, X.; Zhang, Q. Evaluating the Potential of Glycyrrhiza Uralensis (Licorice) in Treating Alcoholic Liver Injury: A Network Pharmacology and Molecular Docking Analysis Approach. Processes 2022, 10, 1808. [Google Scholar] [CrossRef]
  27. Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. AutoDock4 and AutoDockTools4: Automated Docking with Selective Receptor Flexibility. J. Comput Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef] [Green Version]
  28. Alzarea, S.I.; Qasim, S.; Uttra, A.M.; Khan, Y.H.; Aljoufi, F.A.; Ahmed, S.R.; Alanazi, M.; Malhi, T.H. Network Pharmacology and Molecular Docking Based Prediction of Mechanism of Pharmacological Attributes of Glutinol. Processes 2022, 10, 1492. [Google Scholar] [CrossRef]
  29. Pires, D.E.V.; Blundell, T.L.; Ascher, D.B. PkCSM: Predicting Small-Molecule Pharmacokinetic and Toxicity Properties Using Graph-Based Signatures. J. Med. Chem. 2015, 58, 4066–4072. [Google Scholar] [CrossRef]
  30. Gilson, M.K.; Liu, T.; Baitaluk, M.; Nicola, G.; Hwang, L.; Chong, J. BindingDB in 2015: A Public Database for Medicinal Chemistry, Computational Chemistry and Systems Pharmacology. Nucleic Acids Res. 2016, 44, D1045–D1053. [Google Scholar] [CrossRef]
  31. Szklarczyk, D.; Franceschini, A.; Wyder, S.; Forslund, K.; Heller, D.; Huerta-Cepas, J.; Simonovic, M.; Roth, A.; Santos, A.; Tsafou, K.P.; et al. STRING V10: Protein-Protein Interaction Networks, Integrated over the Tree of Life. Nucleic Acids Res. 2015, 43, D447–D452. [Google Scholar] [CrossRef] [Green Version]
  32. Sherman, B.T.; Hao, M.; Qiu, J.; Jiao, X.; Baseler, M.W.; Lane, H.C.; Imamichi, T.; Chang, W. DAVID: A Web Server for Functional Enrichment Analysis and Functional Annotation of Gene Lists (2021 Update). Nucleic Acids Res. 2022, 50, W216–W221. [Google Scholar] [CrossRef]
  33. Oh, K.-K.; Adnan, M.; Cho, D.-H. The Promising Mechanisms of Low Molecular Weight Compounds of Panax Ginseng C.A. Meyer in Alleviating COVID-19: A Network Pharmacology Analysis. Processes 2022, 10, 333. [Google Scholar] [CrossRef]
  34. Keiser, M.J.; Roth, B.L.; Armbruster, B.N.; Ernsberger, P.; Irwin, J.J.; Shoichet, B.K. Relating Protein Pharmacology by Ligand Chemistry. Nat. Biotechnol. 2007, 25, 197–206. [Google Scholar] [CrossRef] [Green Version]
  35. Kim, M.; Kim, Y.B. Uncovering Quercetin’s Effects against Influenza A Virus Using Network Pharmacology and Molecular Docking. Processes 2021, 9, 1627. [Google Scholar] [CrossRef]
  36. Lim, D.-W.; Kim, D.-H.; Yu, G.-R.; Park, W.-H.; Kim, J.-E. Verification of the Potential Targets of the Herbal Prescription Sochehwan for Drug Repurposing Processes as Deduced by Network Pharmacology. Processes 2021, 9, 2034. [Google Scholar] [CrossRef]
  37. Wang, J.F.; Zhou, H.; Han, L.Y.; Chen, X.; Chen, Y.Z.; Cao, Z.W. Traditional Chinese Medicine Information Database. Clinical Pharmacol. Therap. 2005, 78, 92–93. [Google Scholar] [CrossRef]
  38. Zhang, S.; Ge, Q.; Chen, L.; Chen, K. Studies of the Anti-Diabetic Mechanism of Pueraria Lobata Based on Metabolomics and Network Pharmacology. Processes 2021, 9, 1245. [Google Scholar] [CrossRef]
  39. Wishart, D.S.; Feunang, Y.D.; Guo, A.C.; Lo, E.J.; Marcu, A.; Grant, J.R.; Sajed, T.; Johnson, D.; Li, C.; Sayeeda, Z.; et al. DrugBank 5.0: A Major Update to the DrugBank Database for 2018. Nucleic Acids Res. 2018, 46, D1074–D1082. [Google Scholar] [CrossRef]
  40. Oh, K.-K.; Adnan, M.; Cho, D.-H. Network Pharmacology Study to Interpret Signaling Pathways of Ilex Cornuta Leaves against Obesity. Processes 2021, 9, 1106. [Google Scholar] [CrossRef]
  41. Pujol, A.; Mosca, R.; Farrés, J.; Aloy, P. Unveiling the Role of Network and Systems Biology in Drug Discovery. Trends Pharmacol. Sci. 2010, 31, 115–123. [Google Scholar] [CrossRef] [PubMed]
  42. Jiménez-Luna, J.; Grisoni, F.; Schneider, G. Drug Discovery with Explainable Artificial Intelligence. Nat. Mach. Intell. 2020, 2, 573–584. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tang, J. Special Issue on Network Pharmacology Modeling for Drug Discovery. Processes 2023, 11, 1988. https://doi.org/10.3390/pr11071988

AMA Style

Tang J. Special Issue on Network Pharmacology Modeling for Drug Discovery. Processes. 2023; 11(7):1988. https://doi.org/10.3390/pr11071988

Chicago/Turabian Style

Tang, Jing. 2023. "Special Issue on Network Pharmacology Modeling for Drug Discovery" Processes 11, no. 7: 1988. https://doi.org/10.3390/pr11071988

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop