Next Article in Journal
Development and Validation of an HPLC-UV Method for the Quantification of Acyclovir and Ganciclovir in the Plasma of Pediatric Immunocompromised Patients
Previous Article in Journal
Potential Rheumatoid Arthritis-Associated Interstitial Lung Disease Treatment and Computational Approach for Future Drug Development
Previous Article in Special Issue
Structure-Based In Silico Screening of Marine Phlorotannins for Potential Walrus Calicivirus Inhibitor
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue “Computer-Aided Drug Discovery and Treatment”

Department of Medicine, Hadassah Medical Center, Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
Int. J. Mol. Sci. 2024, 25(5), 2683; https://doi.org/10.3390/ijms25052683
Submission received: 24 January 2024 / Accepted: 21 February 2024 / Published: 26 February 2024
(This article belongs to the Special Issue Computer-Aided Drug Discovery and Treatment)
This Special Issue aims to highlight some of the latest developments in drug discovery. Computer-Aided Drug Discovery and Treatment (CADDT) is a multidisciplinary approach that combines bioinformatics, chemoinformatics, structural biology, and artificial intelligence (AI) [1]. CADDT has proven to be instrumental in navigating diseases’ intricate landscapes by facilitating drug development, optimizing treatment strategies, and improving the patients’ outcomes [2].
CADDT is fundamentally based on virtual screening [3]. The molecular binding affinity is predicted using computational models of target proteins. Drug discovery traditionally involves synthesizing and testing many chemical compounds, which is a time-consuming and resource-intensive process. Through virtual screening, researchers can computationally evaluate potential drug candidates, prioritizing those that are likely to exhibit the desired therapeutic effects. Reducing the number of compounds that need to be synthesized and experimentally validated expedites the drug discovery timeline. CADDT allows the design of molecules with specific properties that make them effective against a target protein [4,5].
In structural biology and molecular dynamics simulations, X-ray crystallography, NMR spectroscopy, and cryo-electron microscopy are essential for determining the biomolecule’s three-dimensional structures [6]. The structures serve as a basis for understanding how the drugs interact with their target proteins, facilitating the development of more effective and selective therapeutics. We can better understand the biological molecules’ dynamic behavior by simulating molecular dynamics. Through the computational modeling of atoms and molecules’ movements over time, the proteins’ flexibility and conformational changes provide insights into the drug molecules’ binding mechanisms to optimize drug designs and predict drug–protein interactions under various conditions [7].
The interdisciplinary field of chemoinformatics, which combines chemistry and informatics, plays a critical role in CADDT [8]. Computational methods are used to analyze chemical data, extract meaningful patterns, and predict chemical properties. They are used to identify potential drug candidates for virtual screening, similarity searches, and chemical database analysis [3]. As part of chemoinformatics, ligand-based drug design creates new molecules based on existing ligands. A quantitative structure–activity relationship (QSAR) model enables correlation between the compounds’ chemical structure and biological activity, which can be used to design new molecules with greater potency and selectivity [9].
CADDT is being enhanced using artificial intelligence and machine learning [3]. They can extract knowledge and predict drug discovery and treatment outcomes by analyzing vast datasets. They can identify challenging patterns by analyzing complex biological and chemical data. They allow one to predict drug–drug interactions, analyze patient-specific data, and suggest novel drug targets based on large-scale biological and chemical information. By continuously exposing the models to more data, they become more accurate, identifying new drug candidates and optimizing individualized treatment strategies [10].
CADDT can optimize the pharmacokinetics and pharmacodynamics of drugs by providing information about their absorption, distribution, metabolism, and excretion in the body [11,12]. It is crucial to have this knowledge to design drug formulations that have enhanced efficacy, minimize the side effects, and improve patients’ compliance. Incorporating PK/PD models allows researchers to simulate different dosing regimens, predict drug concentrations at target sites, and optimize treatment protocols for the maximum therapeutic benefit. The existing therapeutics are rendered more effective by refining their administration strategies [13].
A CADDT approach contributes to personalized medicine and patient-specific treatment. By analyzing genetic variations, lifestyle factors, and other patient-specific data, CADDT can identify optimal drug regimens that are likely to be effective with minimal side effects [14]. A predictive model is used in CADDT to identify the patients that are more likely to respond positively to a particular treatment. By stratifying these patients according to their molecular profiles, more targeted and personalized treatments can be delivered [15].
Using Constrained Disorder Principle (CDP)-based systems to improve CADDT: Second-generation AI systems that use CDP-based variability signatures improve CADDT’s accuracy and personalize its output [16]. CDP-based systems account for the biological noise that characterizes all biological systems. These systems focus on the patients’ clinical outcomes as an output for the algorithms and continuously implement signs of biological variability into the algorithm to improve drugs’ efficacy. These systems improve the response to chronic medications, overcome drug tolerance, and reduce the number of side effects, thereby improving patients’ adherence [17]. The option for CDP-based drug design was suggested to develop a variability-based molecular structure.
Ethical Considerations and Challenges: The accuracy of computational models, the integration of several complex data sources, and the validity of virtual predictions through experimental means remain ongoing concerns [18,19]. Computational models are only as reliable as the data used to train them are, which can introduce biases and limitations. In addition to data privacy, transparency, and responsible AI use in healthcare, the ethical considerations must be carefully considered [20].
In summary, CADDT incorporates computational methods and cutting-edge structural biology technologies. By accelerating drug discovery, it improves the personalization and effectiveness of treatments. In recent years, the approach to drug development has been transformed by virtual screening, rational drug design, structural biology, chemoinformatics, and artificial intelligence, which optimize the identification of potential drug candidates and the creation of treatment strategies tailored to each patient. Emerging immersive technologies and quantum computing may further improve the capabilities and accuracy of CADDT.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Niazi, S.K.; Mariam, Z. Computer-Aided Drug Design and Drug Discovery: A Prospective Analysis. Pharmaceuticals 2024, 17, 22. [Google Scholar] [CrossRef] [PubMed]
  2. Villalona-Calero, M.A.; Malhotra, J.; Chung, V.; Xing, Y.; Gray, S.W.; Hampel, H.; Gruber, S.; McDonnell, K. Integrating Early-Stage Drug Development with Clinical Networks; Challenges and Opportunities: The City of Hope Developing Experience. J. Clin. Med. 2023, 12, 4061. [Google Scholar] [CrossRef]
  3. Oliveira, T.A.d.; Silva, M.P.d.; Maia, E.H.B.; Silva, A.M.d.; Taranto, A.G. Virtual Screening Algorithms in Drug Discovery: A Review Focused on Machine and Deep Learning Methods. Drugs Drug Candidates 2023, 2, 311–334. [Google Scholar] [CrossRef]
  4. Gimeno, A.; Ojeda-Montes, M.J.; Tomás-Hernández, S.; Cereto-Massagué, A.; Beltrán-Debón, R.; Mulero, M.; Pujadas, G.; Garcia-Vallvé, S. The Light and Dark Sides of Virtual Screening: What Is There to Know? Int. J. Mol. Sci. 2019, 20, 1375. [Google Scholar] [CrossRef] [PubMed]
  5. Dai, R.; Gao, H.; Su, R. Computer-aided drug design for virtual-screening and active-predicting of main protease (Mpro) inhibitors against SARS-CoV-2. Front. Pharmacol. 2023, 14, 1288363. [Google Scholar] [CrossRef]
  6. Cerofolini, L.; Fragai, M.; Ravera, E.; Diebolder, C.A.; Renault, L.; Calderone, V. Integrative Approaches in Structural Biology: A More Complete Picture from the Combination of Individual Techniques. Biomolecules 2019, 9, 370. [Google Scholar] [CrossRef] [PubMed]
  7. Adelusi, T.I.; Oyedele, A.-Q.K.; Boyenle, I.D.; Ogunlana, A.T.; Adeyemi, R.O.; Ukachi, C.D.; Idris, M.O.; Olaoba, O.T.; Adedotun, I.O.; Kolawole, O.E.; et al. Molecular modeling in drug discovery. Inform. Med. Unlocked 2022, 29, 100880. [Google Scholar] [CrossRef]
  8. Zdrazil, B.; Guha, R. Diversifying cheminformatics. J. Cheminform. 2022, 14, 25. [Google Scholar] [CrossRef] [PubMed]
  9. Kausar, S.; Falcao, A.O. An automated framework for QSAR model building. J. Cheminform. 2018, 10, 1. [Google Scholar] [CrossRef] [PubMed]
  10. Niazi, S.K. The Coming of Age of AI/ML in Drug Discovery, Development, Clinical Testing, and Manufacturing: The FDA Perspectives. Drug Des. Dev. Ther. 2023, 17, 2691–2725. [Google Scholar] [CrossRef]
  11. Pereira, L.C.; Fátima, M.A.; Santos, V.V.; Brandão, C.M.; Alves, I.A.; Azeredo, F.J. Pharmacokinetic/Pharmacodynamic Modeling and Application in Antibacterial and Antifungal Pharmacotherapy: A Narrative Review. Antibiotics 2022, 11, 986. [Google Scholar] [CrossRef] [PubMed]
  12. Zou, H.; Banerjee, P.; Leung, S.S.Y.; Yan, X. Application of Pharmacokinetic-Pharmacodynamic Modeling in Drug Delivery: Development and Challenges. Front. Pharmacol. 2020, 11, 997. [Google Scholar] [CrossRef] [PubMed]
  13. Vora, L.K.; Gholap, A.D.; Jetha, K.; Thakur, R.R.S.; Solanki, H.K.; Chavda, V.P. Artificial Intelligence in Pharmaceutical Technology and Drug Delivery Design. Pharmaceutics 2023, 15, 1916. [Google Scholar] [CrossRef] [PubMed]
  14. Collin, C.B.; Gebhardt, T.; Golebiewski, M.; Karaderi, T.; Hillemanns, M.; Khan, F.M.; Salehzadeh-Yazdi, A.; Kirschner, M.; Krobitsch, S.; Eu-Stands Pm, C.; et al. Computational Models for Clinical Applications in Personalized Medicine-Guidelines and Recommendations for Data Integration and Model Validation. J. Pers. Med. 2022, 12, 166. [Google Scholar] [CrossRef]
  15. Ilan, Y. Next-Generation Personalized Medicine: Implementation of Variability Patterns for Overcoming Drug Resistance in Chronic Diseases. J. Pers. Med. 2022, 12, 1303. [Google Scholar] [CrossRef] [PubMed]
  16. Ilan, Y. The constrained disorder principle defines living organisms and provides a method for correcting disturbed biological systems. Comput. Struct. Biotechnol. J. 2022, 20, 6087–6096. [Google Scholar] [CrossRef] [PubMed]
  17. Sigawi, T.; Lehmann, H.; Hurvitz, N.; Ilan, Y. Constrained Disorder Principle-Based Second-Generation Algorithms Implement Quantified Variability Signatures to Improve the Function of Complex Systems. J. Bioinform. Syst. Biol. 2023, 6, 82–89. [Google Scholar] [CrossRef]
  18. Aldoseri, A.; Al-Khalifa, K.N.; Hamouda, A.M. Re-Thinking Data Strategy and Integration for Artificial Intelligence: Concepts, Opportunities, and Challenges. Appl. Sci. 2023, 13, 7082. [Google Scholar] [CrossRef]
  19. Kandi, V.; Vundecode, A.; Godalwar, T.R.; Dasari, S.; Vadakedath, S.; Godishala, V. The current perspectives in clinical research: Computer-assisted drug designing, ethics, and good clinical practice. Borneo J. Pharm. 2022, 5, 161–178. [Google Scholar] [CrossRef]
  20. Ueda, D.; Kakinuma, T.; Fujita, S.; Kamagata, K.; Fushimi, Y.; Ito, R.; Matsui, Y.; Nozaki, T.; Nakaura, T.; Fujima, N.; et al. Fairness of artificial intelligence in healthcare: Review and recommendations. Jpn. J. Radiol. 2024, 42, 3–15. [Google Scholar] [CrossRef] [PubMed]
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

Ilan, Y. Special Issue “Computer-Aided Drug Discovery and Treatment”. Int. J. Mol. Sci. 2024, 25, 2683. https://doi.org/10.3390/ijms25052683

AMA Style

Ilan Y. Special Issue “Computer-Aided Drug Discovery and Treatment”. International Journal of Molecular Sciences. 2024; 25(5):2683. https://doi.org/10.3390/ijms25052683

Chicago/Turabian Style

Ilan, Yaron. 2024. "Special Issue “Computer-Aided Drug Discovery and Treatment”" International Journal of Molecular Sciences 25, no. 5: 2683. https://doi.org/10.3390/ijms25052683

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