Recent Advances in Machine-Lea
Recent Advances in Machine-Lea
Molecular Sciences
Review
Recent Advances in Machine-Learning-Based
Chemoinformatics: A Comprehensive Review
Sarfaraz K. Niazi 1, * and Zamara Mariam 2
Abstract: In modern drug discovery, the combination of chemoinformatics and quantitative structure-
activity relationship (QSAR) modeling has emerged as a formidable alliance, enabling researchers to
harness the vast potential of machine learning (ML) techniques for predictive molecular design and
analysis. This review delves into the fundamental aspects of chemoinformatics, elucidating the intri-
cate nature of chemical data and the crucial role of molecular descriptors in unveiling the underlying
molecular properties. Molecular descriptors, including 2D fingerprints and topological indices, in
conjunction with the structure–activity relationships (SARs), are pivotal in unlocking the pathway
to small-molecule drug discovery. Technical intricacies of developing robust ML-QSAR models,
including feature selection, model validation, and performance evaluation, are discussed herewith.
Various ML algorithms, such as regression analysis and support vector machines, are showcased in
the text for their ability to predict and comprehend the relationships between molecular structures
and biological activities. This review serves as a comprehensive guide for researchers, providing an
understanding of the synergy between chemoinformatics, QSAR, and ML. Due to embracing these
cutting-edge technologies, predictive molecular analysis holds promise for expediting the discovery
of novel therapeutic agents in the pharmaceutical sciences.
2. Exploration of Chemoinformatics
At the intersection of chemistry and informatics, chemoinformatics has emerged
as a potent field in drug discovery, employing inductive learning to predict chemical
phenomena [3,4]. With the exponentially increasing accessibility of chemical data, the
application of ML in chemoinformatics has revolutionized the way researchers now explore,
analyze, and predict the properties and activities of molecules. Compared to a few decades
ago, it has expedited the process by many folds. It focuses on molecular engineering,
molecular manipulation, library design, compound database searching, chemical space
exploration, molecular graph mining, pharmacophore, and scaffold analysis [5–9].
3. Fundamentals of Chemoinformatics
ML models perform prediction tasks based on chemical training data provided in
the form of mathematical equations or a numerical representation. This transformation of
compound structures into machine-learning-ready chemical data involves a complex, mul-
tilayer computational process. The process encompasses descriptor generation, molecular
graphs, fingerprint construction, similarity analysis, chemical space searching, molecular
dynamic simulations, etc. Each layer is interwoven with the preceding layers, significantly
influencing the interpretation of the chemical data by the machine learning models and
enhancing their predictive capabilities.
molecule libraries and have implicitly learned chemical knowledge to create molecules with
combined characteristics of both bioactive natural products and synthetic compounds, such
as DeepMGM. Besides this, generative models have been used for inverse QSAR/QSPR,
which involves generating molecules that meet specific target properties.
The DeepMGM model was trained using drug-like molecules and produced a general
model (g-DeepMGM) capable of generating scaffold-focused libraries. A target-specific
model (t-DeepMGM) for the cannabinoid receptor 2 (CB2) using transfer learning was also
developed. A discriminator was incorporated into DeepMGM for in silico molecular design
and testing. The generated molecule XIE9137 was identified as a potential CB2 allosteric
modulator, highlighting the effectiveness of deep learning in de novo molecular design
and chemical library generation [26,27].
PCA) analysis and a nonlinear PCA variation, surpassing the predictive capabilities of
LASSO regression.
Similarly, the partial least squares (PLS) method has been employed to discern sig-
nificant structural patterns that contribute to the biological activity of a molecule. The
efficiency and accuracy of PLS in combination with unsupervised dimensionality reduction
techniques surpass the approach of explicitly combining unsupervised dimensionality
with multivariate regression. PLS is also widely utilized in the field of 3D-QSAR model-
ing [6,35,36].
Descriptor
Descriptor Type Example
Dimension
The molecule’s atoms, bonds, and
0D Molecular weight, LogP (partition coefficient)
functional groups count
1D Molecular properties in a linear manner Molecular Formula, SMILES & SELFIES
Molecular fingerprint (e.g., Morgan fingerprint),
2D Topological polar surface area (TPSA)
Constitutional descriptors (e.g., atoms, bonds, and rings count)
Molecular shape descriptors (e.g., volume, surface area),
3D Special properties of a molecule
Pharmacophore features
Molecular dynamics descriptors, solvent accessible surface area
Electrostatic potential descriptors with (SASA), radius of gyration (Rg), Time-dependent properties (e.g.,
4D
spatiotemporal aspects dynamic polar surface area (dPSA), time-dependent
dipole moment
These molecular descriptors have been used to select the most relevant properties.
MoDeSus is an ML-based tool used to determine the most informative molecular descriptors
for QSAR studies. Molecular descriptors allow for ligand-based scaffold hopping for hit
and lead optimization, which speeds up the early stages of drug development and has been
used to compare QSAR and QSPR models. Although each type of descriptor plays a vital
role, 3D and 4D descriptors have shown the most significant contribution to identifying
Int. J. Mol. Sci. 2023, 24, 11488 5 of 15
active molecules and potential drug targets. Furthermore, 4D descriptors like CoMFA and
GRID have been used to identify active sites of receptors and characterize interactions
providing insight into the functional properties of small molecules [41–43].
4. QSAR
Based on its physicochemical characteristics, a ligand’s biological response or activ-
ity can be predicted using QSAR analysis [38]. QSAR modeling techniques have been
used to find prospective drug candidates, and these have developed into AI-based QSAR
methods [44]. Modern machine learning approaches can be applied to model QSAR
or quantitative structure–property relationships (QSPR) and create predicative models
based on artificial intelligence [45,46]. Chemoinformatics, QSAR, and machine learning
applications have been used to showcase different structure-based, ligand-based, and
machine-learning-based approaches for drug development. QSAR/QSPR models employ
information on multiple levels, e.g., chemical data, descriptors, molecular graphs, finger-
prints, similarity analyses, and molecular dynamic simulations, to predict the most optimal
properties of a potential drug.
Structure–activity relationship (SAR) analysis investigates how the chemical structure
of a compound relates to its biological activity or properties and plays a crucial role in
exploring potential effects of bioactivity on changes in the chemical structure of drugs.
Quantifying the degree of the structural or chemical similarity between molecules and
extrapolating chemical attributes from molecular similarity are the goals of similarity
analysis [47]. Similarity search mainly aims to identify compounds with similar bioactivity
to a reference molecule but with different chemotypes. This results in scaffold-hopping
derivatives acquired from a reference compound with a novel core structure. Fragment
replacement approaches, fingerprint-based similarity search, pharmacophore matching,
and 3D shape-based similarity search are all examples of computational research for scaffold
hops. Designing molecules of novel scaffolds with increased pharmacological activity and
identical 3D structure but a multimodal deep transformer neural technique for scaffold
hopping aids the distinct 2D structure [48–51].
SARs are also employed in clustering, inter-molecular comparisons, outlier and nov-
elty analysis, diversity quantification, and outlier analysis. Molecular datasets are used
by AIMSim, a unified platform, to carry out similarity-based tasks using binary similarity
metrics and molecular fingerprints [52]. In a study, a tool called the similarity ensemble
approach (SEA) was used to estimate the accuracy of k-nearest neighbors (kNN) QSAR
models constructed for known ligands of each GPCR target individually to discover ac-
tive and inactive molecules [53]. ChemSAR, another tool, offers an integrated web-based
platform for creating SAR classification models, and it is also an online pipelining plat-
form for molecular SAR modeling. For the identification and structural organization of
analog series, SAR analysis, and compound design, various researchers have employed
the SAR Matrix (SARM) concept [54–56]. DeepSARM, which combined deep learning and
generative modeling, was introduced, expanding the scope of the SARM technique. This
improvement made it possible to create target-based analogs by considering the chemical
information from related targets to increase structural uniqueness and variety [57].
The current approach to constructing QSAR models typically involves generating
descriptors for the compounds in the training set, applying descriptor selection algorithms,
and employing statistical fitting methods to build the model. Nevertheless, there have been
investigations into the potential for developing high-quality, interpretable QSAR models
for large and diverse datasets without relying on pre-calculated descriptors. To achieve this
objective, these studies explore using deep learning techniques, specifically long short-term
memory neural networks [58].
machine learning techniques. By following the protocol, QSAR modeling aided by ML and
DL (deep learning) can predict the properties or activities of chemical compounds, toxicity,
and other related physiochemical properties.
GPCR proteins agonist was found. One hundred seventy-six possible antimalarial hits
were found by integrating QSAR and virtual screening [62,63].
SVMs. This integrated approach successfully identified several critical structural features
associated with the desired biological activity, proving SVM helpful in QSAR modeling [86].
5.5. Convolutional Neural Networks, Recurrent Neural Networks, Deep Neural Networks, and
Ensemble Methods
By leveraging the power of neural networks with multiple hidden layers, deep learning
models can effectively learn complex relationships between molecular structures and their
related biological activities. In QSAR, deep learning models, such as convolutional neural
networks (CNNs), recurrent neural networks (RNNs), and deep neural networks (DNNs),
have been utilized to analyze and predict various properties of molecules, including binding
affinity, activity, toxicity, and bioavailability. These models and their ensemble methods
have been applied in QSAR studies to enhance models’ accuracy and predictive power.
CNNs have successfully captured molecular features and patterns from 2D chemical
structures and search spaces. RNNs have been utilized to model sequential data, such as
molecular fingerprints and SMILES strings. DNNs have effectively learned complex rela-
tionships between 3D and 4D molecular descriptors and their respective bioactivity data.
Ensemble methods combining CNN, RNN, and DNN have been employed to improve
prediction performance. These advanced neural network topologies and ensemble methods
have been extensively used in modeling QSAR/QSPR features of small compounds and
conducting pharmacokinetic and pharmacodynamic studies, alongside work in other fields
of chemoinformatics. In particular, CNN’s unmatched capacity for image analysis made
it possible to visualize protein structures as ‘3D images’ with four separate atom-type
channels. These 3D-CNNs were used to compare the microenvironments of amino acids
and predict how mutations might affect the structure of proteins [87]. A Transformer–CNN
architecture was suggested in a study for QSAR modeling and interpretation. Convolu-
tional and element-wise feed-forward layers were used in place of all recurrent units in
the design, and it was discovered that the Transformer-CNN architecture produced good
results for small datasets and converged quickly for QSAR tasks [88].
Recurrent neural networks (RNNs), also known as long short-term memory (LSTM)
networks, are built to recognize both short-term and long-term dependencies in sequential
input. For applications like de novo drug design, where they learn the structural patterns
and rules from SMILES strings to produce novel molecules, LSTM networks have been
used in the context of QSAR. Deep reinforcement learning, variational autoencoders, and
generative adversarial networks (GANs) are other cutting-edge methods used to gener-
ate compounds with precise molecular features while learning latent representations of
molecules. These methods aid in the discovery of novel medication candidates and the
exploration of new chemical territory [27,89–93]. A study that proposed an ensembled
RNN–CNN architecture, DeepCpG, for DNA methylation analysis concluded that com-
bining RNN and CNN improved the performance of the QSAR model [94]. To perform
QSAR analysis utilizing three-dimensional photographs of chemical structures, a brand-
new DL-based method dubbed DeepSnap was created. Without extracting descriptors, this
method may also forecast the potential toxicity of many compounds to different receptors.
To perform QSAR analysis utilizing three-dimensional photographs of chemical structures,
a brand-new DL-based method dubbed DeepSnap was created. Without extracting descrip-
tors, this method may also forecast the potential toxicity of many compounds to different
receptors [95]. CNN, RNN, and deep-learning-based methods have also shown promising
results in QSAR modeling.
screening. External validation is the primary method for evaluating the accuracy of gener-
ated models for the activity prediction of compounds that have not yet been synthesized.
Understanding the variables that control molecular characteristics and creating new com-
pounds with advantageous features depend on QSAR models, which provide information
on the association between activities and structure-based molecular descriptors [96,97].
Even though 3D-QSAR techniques like CoMFA take structural conformation into ac-
count, they are computationally intensive and can introduce errors related to conformation
prediction, ligand orientation, and structural alignment. Consequently, 2D-QSAR models
can provide a viable alternative and sometimes even outperform 3D-QSAR strategies [38].
The creation of verified models for accurate and precise prediction of a compound’s biologi-
cal actions is the ultimate goal of QSAR analysis. When creating QSAR models, metrics like
R2 and QCV2 are generally optimized. The performances of the final models are assessed
using comparable metrics computed on external datasets [98].
A comparative study on 5-nitrofuran-2-yl derivatives as inhibitors of Mycobacterium
tuberculosis H37Rv used statistical parameters, including squared correlation coefficients,
cross-validated correlation coefficients, and Fischer’s value for statistical importance, to
assess the quality of the generated QSAR models. Another study examined several statisti-
cal parameters of 44 published QSAR models for biologically active substances that were
externally validated and presented in academic journals. They concluded that using the
coefficient of determination (R2) alone was insufficient to determine if a QSAR model was
viable. There are benefits and drawbacks to these defined criteria for external validation
that should be considered in QSAR investigations [99].
technique for reading ML models that can be used to comprehend why a model generates
a specific prediction. Counterfactuals are local interpretations that can disclose the contri-
butions of atoms or fragments within particular molecules to identify the most beneficial
or detrimental motifs to consider for future alterations in the context of QSAR models.
Because they resemble counterfactuals, instance-based techniques have been claimed to
provide ‘natural’ model interpretations for researchers. Various approaches to interpre-
tation have been established; however, there are no appropriate standards to assess how
well they apply to the interpretation of QSAR models. An approach known as STONED
(Structure–Topology Optimization for Novel Explanatory Discoveries) is suggested in a
study; it produces molecular counterfactuals for any model. These molecular counterfac-
tuals offer skeletal, molecular structure-based explanations. All molecules produced by
STONED are legitimate substances, so the method does not require training a counterfac-
tual generator. This simplifies the procedure and eliminates the necessity of a generative
counterfactual creator [100,105,106].
8. Conclusions
Applying machine learning techniques in chemoinformatics has contributed signif-
icantly to discovering and designing highly effective drugs. This paper highlights the
significant role of chemoinformatics and ML-based QSAR in drug discovery and develop-
ment. Integrating computational approaches with large-scale data analysis has revolution-
ized the field, enabling efficient exploration of chemical space and predicting biological
activities. Multiple algorithms built for QSAR modeling significantly highlight features
necessary for further designing small molecules. They have demonstrated their effective-
ness in predicting molecular properties and activities, aiding in compound prioritization
and optimization.
The future of chemoinformatics and QSAR modeling holds promising opportunities
for further advancements. Integrating QSAR models with molecular docking techniques
can enhance the accuracy of binding affinity predictions and provide valuable insights into
the interaction between ligands and target proteins. Fragment-based design approaches
can benefit from QSAR models by guiding the selection and optimization of fragments to
develop novel drug candidates. Additionally, integrating QSAR models with de novo drug
generation methods, such as deep learning and generative modeling, opens up possibilities
for computer-assisted design and discovering new molecules with desired properties.
This convergence of QSAR models with molecular docking, fragment-based design,
and de novo drug generation methods holds great potential to accelerate the drug discovery
process, reduce costs, and increase the success rates of identifying novel therapeutic agents.
Continued research and development in this area will undoubtedly pave the way for more
efficient and precise drug design strategies, ultimately benefiting patients and advancing
the field of pharmaceutical sciences.
References
1. Small Molecule Drug Discovery Market Size, Report by 2032. Available online: https://www.precedenceresearch.com/small-
molecule-drug-discovery-market (accessed on 24 May 2023).
2. Brown, F.K. Chapter 35—Chemoinformatics: What is it and How does it Impact Drug Discovery. In Annual Reports in Medicinal
Chemistry; Bristol, J.A., Ed.; Academic Press: New York, NY, USA, 1998; Volume 33, pp. 375–384. [CrossRef]
3. Polanski, J. 4.26-Chemoinformatics. In Comprehensive Chemometrics, 2nd ed.; Elsevier: Amsterdam, The Netherlands, 2020;
pp. 635–676. [CrossRef]
4. Gasteiger, J. Chemoinformatics: Achievements and Challenges, a Personal View. Molecules 2016, 21, 151. [CrossRef]
5. Polanski, J. 4.14-Chemoinformatics. In Comprehensive Chemometrics; Elsevier: Amsterdam, The Netherlands, 2009; pp. 459–506.
[CrossRef]
6. Gasteiger, J. Handbook of Chemoinformatics; Wiley: New York, NY, USA, 2003. [CrossRef]
7. Varnek, A.; Baskin, I.I. Chemoinformatics as a Theoretical Chemistry Discipline. Mol. Inform. 2011, 30, 20–32. [CrossRef]
Int. J. Mol. Sci. 2023, 24, 11488 12 of 15
8. Bajorath, J.; Bajorath, J. (Eds.) Chemoinformatics and Computational Chemical Biology. In Methods in Molecular Biology; Springer
Science+Business Media: Humana Totowa, NJ, USA, 2011. [CrossRef]
9. Kapetanovic, I.M. Computer-aided drug discovery and development (CADDD): In silico-chemico-biological approach. Chem.-Biol.
Interact. 2008, 171, 165–176. [CrossRef]
10. Rutz, A.; Sorokina, M.; Galgonek, J.; Mietchen, D.; Willighagen, E.; Gaudry, A.; Graham, J.G.; Stephan, R.; Page, R.;
Vondrášek, J.; et al. The LOTUS initiative for open natural products research: Knowledge management through Wikidata.
bioRxiv 2021. [CrossRef]
11. Sorokina, M.; Steinbeck, C. Review on natural products databases: Where to find data in 2020. J. Cheminform. 2020, 12, 20.
[CrossRef] [PubMed]
12. Banerjee, P.; Erehman, J.; Gohlke, B.O.; Wilhelm, T.; Preissner, R.; Dunkel, M. Super Natural II—A database of natural products.
Nucleic Acids Res. 2015, 43, D935–D939. [CrossRef]
13. Zeng, X.; Zhang, P.; He, W.; Qin, C.; Chen, S.; Tao, L.; Wang, Y.; Tan, Y.; Gao, D.; Wang, B.; et al. NPASS: Natural product
activity and species source database for natural product research, discovery and tool development. Nucleic Acids Res. 2018, 46,
D1217–D1222. [CrossRef] [PubMed]
14. Wu, Y.; Zhang, F.; Yang, K.; Fang, S.; Bu, D.; Li, H.; Sun, L.; Hu, H.; Gao, K.; Wang, W.; et al. SymMap: An integrative database of
traditional Chinese medicine enhanced by symptom mapping. Nucleic Acids Res. 2019, 47, D1110–D1117. [CrossRef] [PubMed]
15. 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. [CrossRef] [PubMed]
16. Xue, R.; Fang, Z.; Zhang, M.; Yi, Z.; Wen, C.; Shi, T. TCMID: Traditional Chinese medicine integrative database for herb molecular
mechanism analysis. Nucleic Acids Res. 2012, 41, D1089–D1095. [CrossRef]
17. Krenn, M.; Aspuru-Guzik, A.; Nigam, A.; Friederich, P. Self-referencing embedded strings (SELFIES): A 100% robust molecular
string representation. arXiv 2020, 1, 045024. [CrossRef]
18. Engel, T.; Gasteiger, J. (Eds.) Chemoinformatics: Basic Concepts and Methods; Wiley: New York, NY, USA, 2018. Available online:
https://www.wiley.com/en-dk/Chemoinformatics:+Basic+Concepts+and+Methods-p-9783527331093 (accessed on 7 May 2023).
19. Xue, H.; Stanley-Baker, M.; Kong, A.W.K.; Li, H.; Goh, W.W.B. Data considerations for predictive modeling applied to the
discovery of bioactive natural products. Drug Discov. Today 2022, 27, 2235–2243. [CrossRef] [PubMed]
20. Nikolova, N.; Jaworska, J. Approaches to Measure Chemical Similarity—A Review. Qsar Comb. Sci. 2003, 22, 1006–1026.
[CrossRef]
21. Mendez, D.; Gaulton, A.; Bento, A.P.; Chambers, J.; De Veij, M.; Félix, E.; Magariños, M.P.; Mosquera, J.F.; Mutowo, P.;
Nowotka, M.; et al. ChEMBL: Towards direct deposition of bioassay data. Nucleic Acids Res. 2019, 47, D930–D940. [CrossRef]
22. 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. [CrossRef]
23. 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. [CrossRef]
24. Siramshetty, V.B.; Grishagin, I.; Nguyễn, Ð.T.; Peryea, T.; Skovpen, Y.; Stroganov, O.; Katzel, D.; Sheils, T.; Jadhav, A.;
Mathé, E.A.; et al. NCATS Inxight Drugs: A comprehensive and curated portal for translational research. Nucleic Acids Res. 2022,
50, D1307–D1316. [CrossRef]
25. Sussman, J.L.; Lin, D.; Jiang, J.; Manning, N.O.; Prilusky, J.; Ritter, O.; Abola, E.E. Protein Data Bank (PDB): Database of three-
dimensional structural information of biological macromolecules. Acta Crystallogr. Sect. D Biol. Crystallogr. 1998, 54, 1078–1084.
[CrossRef]
26. Moret, M.; Friedrich, L.; Grisoni, F.; Merk, D.; Schneider, G. Generative molecular design in low data regimes. Nat. Mach. Intell.
2020, 2, 171–180. [CrossRef]
27. Segler, M.H.S.; Kogej, T.; Tyrchan, C.; Waller, M.P. Generating Focused Molecule Libraries for Drug Discovery with Recurrent
Neural Networks. ACS Publ. 2017, 4, 120–131. [CrossRef] [PubMed]
28. Haghighatlari, M.; Li, J.; Heidar-Zadeh, F.; Liu, Y.; Guan, X.; Head-Gordon, T. Learning to Make Chemical Predictions: The
Interplay of Feature Representation, Data, and Machine Learning Methods. Chem 2020, 6, 1527–1542. [CrossRef] [PubMed]
29. David, L.; Thakkar, A.; Mercado, R.; Engkvist, O. Molecular representations in AI-driven drug discovery: A review and practical
guide. J. Cheminform. 2020, 12, 56. [CrossRef] [PubMed]
30. Rahman, R.; Dhruba, S.R.; Ghosh, S.; Pal, R. Functional random forest with applications in dose-response predictions. Sci. Rep.
2019, 9, 1628. [CrossRef] [PubMed]
31. Pang, X.; Fu, W.; Wang, J.; Kang, D.; Xu, L.; Zhao, Y.; Liu, A.L.; Du, G.H. Identification of Estrogen Receptor α Antagonists from
Natural Products via In Vitro and In Silico Approaches. Oxid. Med. Cell. Longev. 2018, 2018, 6040149. [CrossRef] [PubMed]
32. Feinberg, E.N.; Joshi, E.; Pande, V.S.; Cheng, A. Improvement in ADMET Prediction with Multitask Deep Featurization. J. Med.
Chem. 2020, 63, 8835–8848. [CrossRef]
33. Wei, Y.; Li, W.; Du, T.; Hong, Z.; Lin, J. Targeting HIV/HCV Coinfection Using a Machine Learning-Based Multiple Quantitative
Structure-Activity Relationships (Multiple QSAR) Method. Int. J. Mol. Sci. 2019, 20, 3572. [CrossRef] [PubMed]
34. Xiong, J.; Xiong, Z.; Chen, K.; Jiang, H.; Zheng, M. Graph neural networks for automated de novo drug design. Drug Discov.
Today 2021, 26, 1382–1393. [CrossRef]
35. Kubinyi, H. Evolutionary variable selection in regression and PLS analyses. J. Chemom. 1996, 10, 119–133. [CrossRef]
Int. J. Mol. Sci. 2023, 24, 11488 13 of 15
36. Eriksson, L.; Jaworska, J.; Worth, A.; Cronin, M.T.D.; McDowell, R.; Gramatica, P. Methods for reliability and uncertainty
assessment and for applicability evaluations of classification- and regression-based QSARs. Environ. Health Perspect. 2003, 111,
1361–1375. [CrossRef]
37. Dehmer, M.; Varmuza, K.; Bonchev, D. Statistical Modelling of Molecular Descriptors in QSAR/QSPR; Wiley-VCH Verlag
GmbH & Co. KGaA: Weinheim, Germany, 2012. [CrossRef]
38. Lo, Y.; Rensi, S.E.; Torng, W.; Altman, R.B. Machine learning in chemoinformatics and drug discovery. Drug Discov. Today 2018, 23,
1538–1546. [CrossRef]
39. Chandrasekaran, B.; Abed, S.N.; Al-Attraqchi, O.; Kuche, K.; Tekade, R.K. Computer-Aided Prediction of Pharmacokinetic (ADMET)
Properties; Elsevier: Amsterdam, The Netherlands, 2018; pp. 731–755. [CrossRef]
40. Engel, T. Basic Overview of Chemoinformatics. J. Chem. Inf. Model. 2006, 46, 2267–2277. [CrossRef]
41. Ash, J.; Fourches, D. Characterizing the Chemical Space of ERK2 Kinase Inhibitors Using Descriptors Computed from Molecular
Dynamics Trajectories. J. Chem. Inf. Model. 2017, 57, 1286–1299. [CrossRef] [PubMed]
42. Concepts and Experimental Protocols of Modelling and Informatics in Drug Design. ScienceDirect. Available online:
https://www.sciencedirect.com/book/9780128205464/concepts-and-experimental-protocols-of-modelling-and-informatics-
in-drug-design (accessed on 24 May 2023).
43. Machine Learning Descriptors for Molecules. ChemIntelligence. 5 January 2021. Available online: https://chemintelligence.com/
blog/machine-learning-descriptors-molecules (accessed on 14 May 2023).
44. Jiménez-Luna, J.; Grisoni, F.; Weskamp, N.; Schneider, G. Artificial intelligence in drug discovery: Recent advances and future
perspectives. Expert Opin. Drug Discov. 2021, 16, 949–959. [CrossRef] [PubMed]
45. Paul, D.; Sanap, G.; Shenoy, S.; Kalyane, D.; Kalia, K.; Tekade, R.K. Artificial intelligence in drug discovery and development.
Drug Discov. Today 2021, 26, 80–93. [CrossRef]
46. Priya, S.; Kumar, A.; Singh, D.B.; Jain, P.; Tripathi, G. Machine learning approaches and their applications in drug discovery and
design. Chem. Biol. Drug Des. 2022, 100, 136–153. [CrossRef]
47. Bajorath, J. Molecular Similarity Concepts for Informatics Applications. Methods Mol. Biol. 2017, 1526, 231–245. [CrossRef]
48. Sun, H.; Tawa, G.J.; Wallqvist, A. Classification of scaffold-hopping approaches. Drug Discov. Today 2012, 17, 310–324. [CrossRef]
49. Zheng, S.; Lei, Z.; Haitao, A.; Chen, H.; Deng, D.; Yang, Y. Deep scaffold hopping with multimodal transformer neural networks.
J. Cheminform. 2021, 13, 87. [CrossRef] [PubMed]
50. Jenkins, J.L.; Glick, M.; Davies, J. A 3D Similarity Method for Scaffold Hopping from Known Drugs or Natural Ligands to New
Chemotypes. J. Med. Chem. 2004, 47, 6144–6159. [CrossRef]
51. Grisoni, F.; Merk, D.; Consonni, V.; Hiss, J.A.; Tagliabue, S.G.; Todeschini, R.; Schneider, G. Scaffold hopping from natural
products to synthetic mimetics by holistic molecular similarity. Commun. Chem. 2018, 1, 44. [CrossRef]
52. Bhattacharjee, H.; Burns, J.; Vlachos, D.G. AIMSim: An accessible cheminformatics platform for similarity operations on chemicals
datasets. Comput. Phys. Commun. 2023, 283, 108579. [CrossRef]
53. Luo, M.; Wang, X.S.; Tropsha, A. Comparative Analysis of QSAR-based vs. Chemical Similarity Based Predictors of GPCRs
Binding Affinity. Mol. Inform. 2016, 35, 36–41. [CrossRef] [PubMed]
54. Dong, J.; Yao, Z.; Zhu, M.; Wang, N.; Lu, B.; Chen, A.F.; Lu, A.; Miao, H.; Zeng, W.; Cao, D. ChemSAR: An online pipelining
platform for molecular SAR modeling. J. Cheminform. 2017, 9, 27. [CrossRef]
55. Yoshimori, A.; Bajorath, J. The SAR Matrix Method and an Artificially Intelligent Variant for the Identification and Structural
Organization of Analog Series, SAR Analysis, and Compound Design. Mol. Inform. 2020, 39, 2000045. [CrossRef] [PubMed]
56. Hu, H.; Bajorath, J. Systematic assessment of structure-promiscuity relationships between different types of kinase inhibitors.
Bioorganic. Med. Chem. 2021, 41, 116226. [CrossRef] [PubMed]
57. Yoshimori, A.; Hu, H.; Bajorath, J. Adapting the DeepSARM approach for dual-target ligand design. J. Comput.-Aided Mol. Des.
2021, 35, 587–600. [CrossRef] [PubMed]
58. Chakravarti, S.K.; Alla, S.R.M. Descriptor Free QSAR Modeling Using Deep Learning with Long Short-Term Memory Neural
Networks. Front. Artif. Intell. 2019, 2, 17. [CrossRef]
59. Ponzoni, I.; Sebastián-Pérez, V.; Requena-Triguero, C.; Roca, C.P.; Martínez, M.J.; Cravero, F.; Díaz MP, M.; Páez, J.A.; Arrayás, R.G.;
Adrio, J.; et al. Hybridizing Feature Selection and Feature Learning Approaches in QSAR Modeling for Drug Discovery. Sci. Rep.
2017, 7, 2403. [CrossRef]
60. Tropsha, A. Best Practices for QSAR Model Development, Validation, and Exploitation. Mol. Inform. 2010, 29, 476–488. [CrossRef]
61. Kumar, N.; Acharya, V. Machine intelligence-driven framework for optimized hit selection in virtual screening. J. Cheminform.
2022, 14, 48. [CrossRef]
62. Tsou, L.K.; Yeh, S.H.; Ueng, S.; Chang, C.; Song, J.; Wu, M.; Chang, H.T.; Chen, S.; Shih, C.; Chen, C.; et al. Comparative study
between deep learning and QSAR classifications for TNBC inhibitors and novel GPCR agonist discovery. Sci. Rep. 2020, 10, 16771.
[CrossRef] [PubMed]
63. Neves, B.M.; Braga, R.C.; Melo-Filho, C.C.; Moreira-Filho, J.T.; Muratov, E.N.; Andrade, C.H. QSAR-Based Virtual Screening:
Advances and Applications in Drug Discovery. Front. Pharmacol. 2018, 9, 1275. [CrossRef] [PubMed]
64. Duchowicz, P.R. Linear Regression QSAR Models for Polo-Like Kinase-1 Inhibitors. Cells 2018, 7, 13. [CrossRef] [PubMed]
65. Cardoso-Silva, J.; Papageorgiou, L.G.; Tsoka, S. Network-based piecewise linear regression for QSAR modelling. J. Comput.-Aided
Mol. Des. Vol. 2019, 33, 831–844. [CrossRef]
Int. J. Mol. Sci. 2023, 24, 11488 14 of 15
66. Dudek, A.Z.; Arodz, T.; Galvez, J. Computational Methods in Developing Quantitative Structure-Activity Relationships (QSAR):
A Review. Comb. Chem. High Throughput Screen. 2006, 9, 213–228. [CrossRef]
67. Raevsky, O.A.; Sapegin, A.; Zefirov, N.S. The QSAR Discriminant-Regression Model. Quant. Struct.-Act. Relatsh. 1994, 13, 412–418.
[CrossRef]
68. Doreswamy; Vastrad, B. Predictive Comparative Qsar Analysis of as 5-Nitrofuran-2-Yl Derivatives Myco Bacterium Tuberculosis
H37RV Inhibitors. Healthc. Inform. Int. J. 2013, 2, 47–62. [CrossRef]
69. Ajmani, S.; Jadhav, K.M.; Kulkarni, S.A. Three-Dimensional QSAR Using the k-Nearest Neighbor Method and Its Interpretation.
J. Chem. Inf. Model. 2006, 46, 24–31. [CrossRef]
70. 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. [CrossRef]
71. Raj, N.; Jain, S. 3d QSAR Studies in Conjunction With k-Nearest Neighbor Molecular Field Analysis (k-NN-MFA) on
a Series of ResearchGate. 2011. Available online: https://www.researchgate.net/publication/294708142_3d_QSAR_
studies_in_conjunction_with_k-nearest_neighbor_molecular_field_analysis_k-NN-MFA_on_a_series_of_substituted_2-
phenyl-benzimidazole_derivatives_as_an_anti_allergic_agents (accessed on 24 May 2023).
72. Asikainen, A.H.; Ruuskanen, J.; Tuppurainen, K.A. Consensus kNN QSAR: A versatile method for predicting the estrogenic
activity of organic compounds in silico. A comparative study with five estrogen receptors and a large, diverse set of ligands.
Environ. Sci. Technol. 2004, 38, 6724–6729. [CrossRef]
73. Nigsch, F.; Bender, A.; Van Buuren, B.N.; Tissen, J.; Nigsch, E.A.; Mitchell, J.C. Melting Point Prediction Employing k-Nearest
Neighbor Algorithms and Genetic Parameter Optimization. J. Chem. Inf. Model. 2006, 46, 2412–2422. [CrossRef] [PubMed]
74. Poroikov, V.V.; Filimonov, D.A.; Borodina, Y.V.; Lagunin, A.A.; Kos, A. Robustness of biological activity spectra predicting by
computer program PASS for noncongeneric sets of chemical compounds. J. Chem. Inf. Comput. Sci. 2000, 40, 1349–1355. [CrossRef]
75. Chen, B.; Sheridan, R.P.; Hornak, V.; Voigt, J.H. Comparison of random forest and Pipeline Pilot Naïve Bayes in prospective QSAR
predictions. J. Chem. Inf. Model. 2012, 52, 792–803. [CrossRef] [PubMed]
76. Kupervasser, O. Quantitative Structure-Activity Relationship Modeling and Bayesian Networks: Optimality of Naive Bayes Model;
IntechOpen: Rijeka, Croatia, 2019. [CrossRef]
77. Eklund, M.; Norinder, U.; Boyer, S.; Carlsson, L. Choosing Feature Selection and Learning Algorithms in QSAR. J. Chem. Inf.
Model. 2014, 54, 837–843. [CrossRef]
78. Bender, A.; Jenkins, J.L.; Glick, M.; Deng, Z.; Nettles, J.H.; Davies, J.W. “Bayes affinity fingerprints” improve retrieval rates in
virtual screening and define orthogonal bioactivity space: When are multitarget drugs a feasible concept? J. Chem. Inf. Model.
2006, 46, 2445–2456. [CrossRef] [PubMed]
79. Keyvanpour, M.R.; Shirzad, M.B. An Analysis of QSAR Research Based on Machine Learning Concepts. Curr. Drug Discov.
Technol. 2021, 18, 17–30. [CrossRef] [PubMed]
80. Bugeac, C.A.; Ancuceanu, R.; Dinu, M. QSAR Models for Active Substances against Pseudomonas aeruginosa Using Disk-
Diffusion Test Data. Molecules 2021, 26, 1734. [CrossRef]
81. Darnag, R.; Schmitzer, A.R.; Belmiloud, Y.; Villemin, D.; Jarid, A.; Chait, A.; Seyagh, M.; Cherqaoui, D. QSAR Studies of HEPT
Derivatives Using Support Vector Machines. Qsar Comb. Sci. 2009, 28, 709–718. [CrossRef]
82. Niu, B.; Lu, W.; Yang, S.; Cai, Y.; Li, G. Support vector machine for SAR/QSAR of phenethyl-amines. Acta Pharmacol. Sin. 2007, 28,
1075–1086. [CrossRef]
83. Wu, Z.; Zhu, M.; Kang, Y.; Leung, E.L.; Lei, T.; Shen, C.; Jiang, D.; Wang, Z.; Cao, D.; Hou, T. Do we need different machine
learning algorithms for QSAR modeling? A comprehensive assessment of 16 machine learning algorithms on 14 QSAR data sets.
Brief. Bioinform. 2021, 22, bbaa321. [CrossRef]
84. Alvarsson, J.; Lampa, S.; Schaal, W.; Andersson, C.; Wikberg, J.E.S.; Spjuth, O. Large-scale ligand-based predictive modelling
using support vector machines. J. Cheminform. 2016, 8, 39. [CrossRef]
85. Liu, H.X.; Zhang, R.S.; Yao, X.J.; Liu, M.C.; Hu, Z.D.; Fan, B.T. QSAR study of ethyl 2-[(3-methyl-2,5-dioxo(3-pyrrolinyl))amino]-4-
(trifluoromethyl) pyrimidine-5-carboxylate: An inhibitor of AP-1 and NF-kappa B mediated gene expression based on support
vector machines. J. Chem. Inf. Comput. Sci. 2003, 43, 1288–1296. [CrossRef] [PubMed]
86. Nekoei, M.; Mohammadhosseini, M.; Pourbasheer, E. QSAR study of VEGFR-2 inhibitors by using genetic algorithm-multiple
linear regressions (GA-MLR) and genetic algorithm-support vector machine (GA-SVM): A comparative approach. Med. Chem.
Res. 2015, 24, 3037–3046. [CrossRef]
87. Torng, W.; Altman, R.B. 3D deep convolutional neural networks for amino acid environment similarity analysis. BMC Bioinform.
2017, 18, 302. [CrossRef] [PubMed]
88. Olivecrona, M.; Blaschke, T.; Engkvist, O.; Chen, H. Molecular de-novo design through deep reinforcement learning. J. Cheminform.
2017, 9, 48. [CrossRef]
89. Graves, A.; Mohamed, A.; Hinton, G.E. Speech Recognition with Deep Recurrent Neural Networks. arXiv (Cornell University).
arxiv 2013, arXiv:1303.5778. [CrossRef]
90. Kingma, D.P. Auto-Encoding Variational Bayes. arXiv 2013, arXiv:1312.6114.
91. Goodfellow, I.J. Generative Adversarial Networks. arXiv 2014, arXiv:1406.2661. [CrossRef]
92. Mnih, V.; Kavukcuoglu, K.; Silver, D.; Rusu, A.; Veness, J.; Bellemare, M.F.; Graves, A.; Riedmiller, M.; Fidjeland, A.K.;
Ostrovski, G.; et al. Human-level control through deep reinforcement learning. Nature 2015, 518, 529–533. [CrossRef]
Int. J. Mol. Sci. 2023, 24, 11488 15 of 15
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.
Reproduced with permission of copyright owner. Further reproduction
prohibited without permission.