Machine Learning Empowering Drug Discovery Applica
Machine Learning Empowering Drug Discovery Applica
Review
Machine Learning Empowering Drug Discovery: Applications,
Opportunities and Challenges
Xin Qi 1, *,† , Yuanchun Zhao 1,† , Zhuang Qi 2 , Siyu Hou 1 and Jiajia Chen 1
1 School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215011, China;
[email protected] (Y.Z.); [email protected] (S.H.); [email protected] (J.C.)
2 School of Software, Shandong University, Jinan 250101, China; [email protected]
* Correspondence: [email protected]; Tel.: +86-0512-68418434
† These authors contributed equally to this work.
Abstract: Drug discovery plays a critical role in advancing human health by developing new medica-
tions and treatments to combat diseases. How to accelerate the pace and reduce the costs of new drug
discovery has long been a key concern for the pharmaceutical industry. Fortunately, by leveraging
advanced algorithms, computational power and biological big data, artificial intelligence (AI) tech-
nology, especially machine learning (ML), holds the promise of making the hunt for new drugs more
efficient. Recently, the Transformer-based models that have achieved revolutionary breakthroughs in
natural language processing have sparked a new era of their applications in drug discovery. Herein,
we introduce the latest applications of ML in drug discovery, highlight the potential of advanced
Transformer-based ML models, and discuss the future prospects and challenges in the field.
1. Introduction
Drug research and development play a vital role in improving human health and
well-being. However, the discovery of a new drug is an extremely complex, expensive and
Citation: Qi, X.; Zhao, Y.; Qi, Z.; Hou, time-consuming process, typically costing approximately USD 2.6 billion [1] and taking
S.; Chen, J. Machine Learning more than 10 years on average [2]. Despite the high investment levels, the approval success
Empowering Drug Discovery: rate of launching a small-molecule drug to market from phase I clinical trial is less than
Applications, Opportunities and 10% [3], highlighting the considerable risk of failure. Therefore, how to reduce the costs
Challenges. Molecules 2024, 29, 903. and accelerate the pace of new drug discovery has emerged as a key concern within the
https://doi.org/10.3390/ pharmaceutical industry.
molecules29040903 The increasing availability of large-scale biomedical data provides tremendous op-
Academic Editor: Shijun Zhong portunities for computational drug discovery, but effectively mining, correlating, and
analyzing these huge amounts of data has become a critical challenge. Fortunately, with
Received: 15 January 2024 the advent of efficient mathematical tools and abundant computational resources, artificial
Revised: 8 February 2024
intelligence (AI) approaches have rapidly developed (Figure 1). As the representative AI
Accepted: 14 February 2024
method, machine learning (ML), empowers machines to learn from existing data by using
Published: 18 February 2024
statistical approaches and make predictions, which can be further classified into supervised,
unsupervised, and reinforcement learnings [4,5]. Deep learning (DL), a subset of ML,
focuses on using multi-layered artificial neural networks (ANNs) structures to simulate
Copyright: © 2024 by the authors.
the neural networks of the human brain for learning data representations, making it more
Licensee MDPI, Basel, Switzerland. powerful and flexible in handling complex and high-dimensional data [6,7]. With the
This article is an open access article advantages of low cost and fast speed, the ML approaches are revolutionizing and strength-
distributed under the terms and ening multiple stages of drug discovery, such as target identification, de novo drug design
conditions of the Creative Commons and drug repurposing. For example, DL-based open-source tools, such as DeepDTAF [8]
Attribution (CC BY) license (https:// and DeepAffinity [9], have been applied to predict the binding affinity of drug–target
creativecommons.org/licenses/by/ interactions (DTIs), making the hunt for new pharmaceuticals more efficient. Accordingly,
4.0/). more and more pharmaceutical giants, such as Sanofi (Paris, France), Merck (Darmstadt,
tools, such as DeepDTAF [8] and DeepAffinity [9], have been applied to predict the
binding affinity of drug–target interactions (DTIs), making the hunt for new pharmaceu-
Molecules 2024, 29, 903 2 of 20
ticals more efficient. Accordingly, more and more pharmaceutical giants, such as Sanofi
(Paris, France), Merck (Darmstadt, Germany), Takeda (Takeda, Japan) and Genentech
(South San Francisco, America), have initiated cooperation with AI companies to develop
Germany),
new drugs.Takeda (Takeda, Japan) and Genentech (South San Francisco, America), have
initiated cooperation with AI companies to develop new drugs.
Figure 1. Introduction diagram of artificial intelligence and its subfields: machine learning and
Figure 1. Introduction diagram of artificial intelligence and its subfields: machine learning and
deep
deep learning.
learning.
Figure 2. Machine learning can be applied in multiple stages of the drug discovery process, mainly
Figure 2. Machine learning can be applied in multiple stages of the drug discovery process, mainly
including drug design, drug screening, drug repurposing and chemical synthesis.
including drug design, drug screening, drug repurposing and chemical synthesis.
Molecules 2024, 29, 903 4 of 20
Table 1. Cont.
Note: DNN, deep neural network; RNN, recurrent neural network; RF, random forest; CNN, convolutional neural
network; XGBoost, eXtreme gradient boosting; GCN, graph convolutional network; GAT, graph attention network;
SVM, support vector machine; NBC, naïve Bayes classifier; ResNet, residual network; GBM, gradient boosting
machines; RL, reinforcement learning; GRU, gated recurrent unit; VAE, variational autoencoder; AAE, adaptive
adversarial autoencoder; GNN, graph neural networks; k-NN, k-nearest neighbor; LightGBM, light gradient
boosting machine; MCTS, Monte Carlo tree search, NTN, neural tensor network; GAN, generative adversarial
network; GCNN, graph convolutional neural network.
on balanced sets with equal numbers of positive and negative pairs was 0.9989, and the
average accuracy of five-fold cross-validation on unbalanced sets with a ratio of positive
and negative pairs of 1:10 was 0.9942 [24]. Comparatively, sequence-based PPI prediction
approaches aim to identify physical interactions between two proteins by leveraging
information from their protein sequences [92]. DNNs provide a robust solution for this
purpose. They are composed of multiple layers of interconnected neurons, allowing them
to automatically extract complex patterns and features from data. For example, DeepPPI
applied DNNs to effectively learn protein representations from common protein descriptors,
thereby contributing to the prediction of PPIs. It can achieve excellent performance on the
S. cerevisiae dataset with an accuracy of 0.925, precision of 0.9438, recall of 0.9056, specificity
of 0.9449, MCC of 0.8508 and area under the curve (AUC) of 0.9743, respectively [27].
Extensive experiments showed that DeepPPI was able to learn the useful features of protein
pairs through a layer-wise abstraction, resulting in better predictive performance than
existing methods on core S. cerevisiae, H. pylori and H. sapien datasets [27]. In addition,
based on Uniprot database, Li et al. [20] developed a DELPHI, a new sequence-based deep
ensemble model for PPI-binding sites’ prediction. Therefore, ML-based approaches have
great potential in enhancing the identification of PPI sites. Compared with sequence-based
approaches, structure-based ones are limited by the scarcity of available protein structures
and the low quality of familiar protein structures [90,93].
de novo drug design methods (e.g., the fragment-based approach) usually have poor drug
metabolism and pharmacokinetics properties and are hard to synthesize due to the com-
plexity and impracticality of compound structures [97,98]. Therefore, there is high demand
for new methods to explore chemical entities that meet the requirements of biological
activity, drug metabolism, pharmacokinetics and synthesis practicality.
Recently, ML-based approaches, especially auto-encoder variants (e.g., the variational
auto-encoder (VAE) and adversarial auto-encoder (AAE)) have gained attention in the field
of de novo drug design. PaccMannRL is an example of these approaches that combines a
hybrid VAE with reinforcement learning for the de novo design of anti-cancer molecules
from transcriptomic data [49]. Similarly, another approach, known as druGAN, utilizes
a deep generative AAE model to generate novel molecules that possess specific anti-
cancer properties [50]. In addition, a Wasserstein GAN and GCN-based model, known as
MedGAN, has been successfully developed to generate novel quinoline-scaffold molecules
from complicated molecular graphs and evaluate drug-related properties [56]. It has been
demonstrated that the MedGAN was able to produce 25% effective molecules, 62% fully
connected, among which 92% are quinoline, 93% are novel, and 95% are unique [56]. To
address the difficulty in synthesizing generated molecules, Coley et al. [51] defined a
synthetic complexity score, namely SCScore, that utilizes precedent reaction knowledge to
train a neural network model for evaluating the level of synthetic complexity. Therefore,
ML-empowering approaches play crucial roles in de novo drug design, revolutionizing the
process of discovering and developing new drugs.
To detect the ADME/T properties of drugs, various evaluation criteria such as hepa-
totoxicity, passing through the blood–brain barrier (BBB), plasma protein binding (PPB)
and cytochrome P450 2D6 (CYP2D6) inhibition are commonly used [101,102]. Accordingly,
there has been growing interest in developing ML-based tools for the prediction of these
criteria. For example, Tian et al. [60] developed a web server called ADMETboost that
utilizes the powerful extreme gradient boosting (XGBoost) model to learn about molecule
features from multiple fingerprints and descriptors, allowing for the accurate prediction
of ADME/T properties, such as Caco2, BBB, CYP2C9 inhibition, CL-Hepa and hERG. It
has been demonstrated that this model can achieve remarkable results in the Therapeutics
Data Commons ADMET benchmark, ranking first in 18 out of 22 tasks and within the top
three in 21 tasks [60]. Similarly, by utilizing more than 13 000 compounds obtained from
the PubChem BioAssay Database, Li et al. [65] proposed a multitask autoencoder DNN
model to predict the inhibitors of five major cytochrome P450 (CYP450) isoforms (1A2, 2C9,
2C19, 2D6 and 3A4). Especially, the multi-task DNN model achieved average prediction
accuracies of 86.4% in 10-fold cross-validation and 88.7% on external test datasets, outper-
forming single-task models, earlier described classifiers and conventional ML methods [65].
Furthermore, the Tox21 Challenge is a collaborative effort aimed at developing predictive
models for toxicity assessment using high-throughput screening data. In this context, Mayr
et al. [64] developed a DL pipeline, DeepTox, for toxicity prediction. It outperformed
all other computational methods (e.g., naïve Bayes, random forest and support vector
machine) in 10 out of 15 cases in the Tox21 Challenge [64]. Therefore, ML algorithms have
made significant progress in predicting the ADME/T properties of drugs, contributing to
guiding drug safety assessment and preclinical research.
methods integrate information from different biological networks to improve the predictive
accuracy of drug–disease relationships. For example, Doshi et al. [74] developed a graph
neural network model called GDRnet for drug repurposing, which can efficiently screen
existing drugs in the database and predict their unknown therapeutic effects by evaluating
the scores of drug–disease pairs. Therefore, ML technology holds significant promise in the
field of drug repurposing, providing strong support for accelerating drug discovery.
Molecules 2024, 29, 903 to predict the optimal reaction conditions for various types of reactions. This model was 11 of 21
trained using a vast dataset of nearly 10 million entries extracted from the Reaxys database
and can effectively predict the ideal catalyst, solvent, reagent, and temperature for a given
reaction, facilitating the optimization of reaction conditions [84]. Therefore, the utilization
identifying
of ML-based optimal
modelsreaction conditions,
can assist thereby
in predicting holding
reaction types,great potential
accelerating indiscovery
the improving of the
efficiency of chemical
new chemical synthesis
molecules, processes.optimal reaction conditions, thereby holding
and identifying
great potential in improving the efficiency of chemical synthesis processes.
3. Opportunities for Transformer-Based ML Models in Empowering Drug Discovery
3. Opportunities for Transformer-Based ML Models in Empowering Drug Discovery
The Transformer model, firstly proposed in the paper ‘Attention is All You Need’ by
Vaswani Theet
Transformer model,
al., is a highly firstly proposed
advanced in the paper
DL architecture ‘Attention
utilizing is All Youmechanisms.
self-attention Need’ by
Vaswani et al., is a highly advanced DL architecture utilizing self-attention
As it allows for parallelization and captures long-range dependencies more efficiently mechanisms.
As it allows for parallelization and captures long-range dependencies more efficiently than
than traditional RNN models, the Transformer model has proven to be highly effective in
traditional RNN models, the Transformer model has proven to be highly effective in a wide
a wide range of tasks and has set new benchmarks in the corresponding fields [10,11].
range of tasks and has set new benchmarks in the corresponding fields [10,11]. Given the
Given the advantages of the Transformer, it has emerged as a promising future direction
advantages of the Transformer, it has emerged as a promising future direction of ML in the
offield
MLofindrug
the field of drug
discovery discovery
(Figure 3). (Figure 3).
for target-specific de novo molecular generation. The chemical space of the compounds
generated by cMolGPT closely matches with that of real target-specific ones [114].
5. Concluding Remarks
The research and development of new drugs can contribute to meet the human de-
mand for treating diseases and provide more effective, safer, and more convenient treatment
options. Compared with the traditional strategies of drug discovery, ML-based approaches
have the potential to reduce time and costs, improve safety, and bridge the gap between
drug discovery and drug effectiveness, making them increasingly favored by the pharma-
ceutical industry and academia. In particular, the introduction of chatGPT has sparked
researchers’ growing interest and exploration in leveraging the Transformer model’s NLP
capabilities, particularly its self-attention mechanisms, to accelerate multiple stages of the
drug discovery process, thereby opening up new opportunities for advancements.
However, the current challenges in ML-based models can result in generating false
positives or false negatives, potentially leading to incorrect predictions and resource waste.
Further in vitro and in vivo experiments as well as clinical trials are needed to fully demon-
strate the practicability of ML-based drug discovery and obtain more reliable and accurate
results. Therefore, future research should focus on improving data quality, enhancing the in-
terpretability of ML algorithms, and integrating them with human professional knowledge
to increase the efficacy of drug discovery.
Author Contributions: X.Q. and Y.Z. designed the review; Y.Z. collected the related data; X.Q. and
Y.Z. drafted the manuscript; X.Q., Y.Z., S.H., Z.Q. and J.C. revised the manuscript. All authors have
read and agreed to the published version of the manuscript.
Funding: This work was supported by the National Natural Science Foundation of China (Grant No.
32270705) and the Postgraduate Research & Practice Innovation Program of Jiangsu Province (Grant
No. KYCX23_3344).
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Not applicable.
Conflicts of Interest: The authors declare no conflicts of interest.
Molecules 2024, 29, 903 15 of 20
Abbreviations
AI, artificial intelligence; ML, machine learning; DL, deep learning; ANN, artificial neural network;
NLP, natural language processing; DTI, drug–target interaction; 3D, three-dimensional; DNN, deep
neural network; PPI, protein–protein interaction; GAT, graph attention network; CNN, convolutional
neural network; VAE, variational auto-encoder; AAE, adversarial auto-encoder; RNN, recurrent
neural network; RF, random forest; XGBoost, eXtreme gradient boosting; GCN, graph convolutional
network; SVM, support vector machine; NBC, naïve Bayes classifier; NTN, neural tensor network;
GAN, generative adversarial network; GCNN, graph convolutional neural network; ResNet, residual
network; GBM, gradient boosting machines; RL, reinforcement learning; GRU, gated recurrent unit;
GNN, graph neural networks; k-NN, k-nearest neighbor; LightGBM, light gradient boosting machine;
MCTS, Monte Carlo tree search, NTN, neural tensor network; GAN, generative adversarial network;
GCNN, graph convolutional neural network; ADME/T, absorption, distribution, metabolism, and
excretion and toxicity; BBB, blood–brain barrier; PPB, plasma protein binding, CYP2D6, cytochrome
P450 2D6; XGBoost, extreme gradient boosting; CYP450, cytochrome P450;MCTS, Monte Carlo Tree
Search; GPT, Generative Pre-Training Transformer; BERT, bidirectional encoder representations from
transformers; SC2, Challenge to Predict Aqueous Solubility; MCC, Matthews’ Correlation Coefficient;
AUC, area under the curve; AUPR, area under precision-recall; PDB, Protein Data Bank; QED,
quantitative estimate of drug-likeness; SA, synthetic accessibility;NP, natural products; LIME, Local
Interpretable Model-agnostic Explanations; SHAP, SHapley Additive exPlanations.
References
1. DiMasi, J.A.; Grabowski, H.G.; Hansen, R.W. Innovation in the pharmaceutical industry: New estimates of R&D costs. J. Health
Econ. 2016, 47, 20–33. [CrossRef] [PubMed]
2. Ashburn, T.T.; Thor, K.B. Drug repositioning: Identifying and developing new uses for existing drugs. Nat. Rev. Drug Discov.
2004, 3, 673–683. [CrossRef]
3. Dowden, H.; Munro, J. Trends in clinical success rates and therapeutic focus. Nat. Rev. Drug Discov. 2019, 18, 495–496. [CrossRef]
[PubMed]
4. Deng, J.; Yang, Z.; Ojima, I.; Samaras, D.; Wang, F. Artificial intelligence in drug discovery: Applications and techniques. Brief.
Bioinform 2022, 23, bbab430. [CrossRef] [PubMed]
5. Mak, K.K.; Pichika, M.R. Artificial intelligence in drug development: Present status and future prospects. Drug Discov. Today 2019,
24, 773–780. [CrossRef]
6. 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]
7. Chen, H.; Engkvist, O.; Wang, Y.; Olivecrona, M.; Blaschke, T. The rise of deep learning in drug discovery. Drug Discov. Today
2018, 23, 1241–1250. [CrossRef]
8. Wang, K.; Zhou, R.; Li, Y.; Li, M. DeepDTAF: A deep learning method to predict protein-ligand binding affinity. Brief. Bioinform.
2021, 22, bbab072. [CrossRef]
9. Karimi, M.; Wu, D.; Wang, Z.; Shen, Y. DeepAffinity: Interpretable deep learning of compound-protein affinity through unified
recurrent and convolutional neural networks. Bioinformatics 2019, 35, 3329–3338. [CrossRef]
10. Zhang, S.; Fan, R.; Liu, Y.; Chen, S.; Liu, Q.; Zeng, W. Applications of transformer-based language models in bioinformatics:
A survey. Bioinform. Adv. 2023, 3, vbad001. [CrossRef]
11. Vaswani, A.; Shazeer, N.; Parmar, N.; Uszkoreit, J.; Jones, L.; Gomez, A.N.; Kaiser, Ł.; Polosukhin, I. Attention is all you need. Adv.
Neural Inf. Process. Syst. 2017, 30, 1–11.
12. Kalakoti, Y.; Yadav, S.; Sundar, D. TransDTI: Transformer-Based Language Models for Estimating DTIs and Building a Drug
Recommendation Workflow. ACS Omega 2022, 7, 2706–2717. [CrossRef] [PubMed]
13. Du, Z.; Su, H.; Wang, W.; Ye, L.; Wei, H.; Peng, Z.; Anishchenko, I.; Baker, D.; Yang, J. The trRosetta server for fast and accurate
protein structure prediction. Nat. Protoc. 2021, 16, 5634–5651. [CrossRef]
14. Senior, A.W.; Evans, R.; Jumper, J.; Kirkpatrick, J.; Sifre, L.; Green, T.; Qin, C.; Žídek, A.; Nelson, A.W.R.; Bridgland, A.; et al.
Improved protein structure prediction using potentials from deep learning. Nature 2020, 577, 706–710. [CrossRef]
15. Zhang, L.; Wang, S.; Hou, J.; Si, D.; Zhu, J.; Cao, R. ComplexQA: A deep graph learning approach for protein complex structure
assessment. Brief. Bioinform. 2023, 24, bbad287. [CrossRef] [PubMed]
16. Brandes, N.; Ofer, D.; Peleg, Y.; Rappoport, N.; Linial, M. ProteinBERT: A universal deep-learning model of protein sequence and
function. Bioinformatics 2022, 38, 2102–2110. [CrossRef]
17. Lin, Z.; Akin, H.; Rao, R.; Hie, B.; Zhu, Z.; Lu, W.; Smetanin, N.; Verkuil, R.; Kabeli, O.; Shmueli, Y.; et al. Evolutionary-scale
prediction of atomic-level protein structure with a language model. Science 2023, 379, 1123–1130. [CrossRef]
Molecules 2024, 29, 903 16 of 20
18. Northey, T.C.; Barešić, A.; Martin, A.C.R. IntPred: A structure-based predictor of protein-protein interaction sites. Bioinformatics
2018, 34, 223–229. [CrossRef]
19. Maheshwari, S.; Brylinski, M. Template-based identification of protein-protein interfaces using eFindSitePPI. Methods 2016, 93,
64–71. [CrossRef]
20. Li, Y.; Golding, G.B.; Ilie, L. DELPHI: Accurate deep ensemble model for protein interaction sites prediction. Bioinformatics 2021,
37, 896–904. [CrossRef]
21. Wang, X.; Zhang, Y.; Yu, B.; Salhi, A.; Chen, R.; Wang, L.; Liu, Z. Prediction of protein-protein interaction sites through eXtreme
gradient boosting with kernel principal component analysis. Comput. Biol. Med. 2021, 134, 104516. [CrossRef]
22. Kang, Y.; Xu, Y.; Wang, X.; Pu, B.; Yang, X.; Rao, Y.; Chen, J. HN-PPISP: A hybrid network based on MLP-Mixer for protein-protein
interaction site prediction. Brief. Bioinform. 2023, 24, bbac480. [CrossRef]
23. Song, B.; Luo, X.; Luo, X.; Liu, Y.; Niu, Z.; Zeng, X. Learning spatial structures of proteins improves protein-protein interaction
prediction. Brief. Bioinform. 2022, 23, bbab558. [CrossRef] [PubMed]
24. Baranwal, M.; Magner, A.; Saldinger, J.; Turali-Emre, E.S.; Elvati, P.; Kozarekar, S.; VanEpps, J.S.; Kotov, N.A.; Violi, A.; Hero, A.O.
Struct2Graph: A graph attention network for structure based predictions of protein-protein interactions. BMC Bioinform. 2022, 23,
370. [CrossRef] [PubMed]
25. Yao, Y.; Du, X.; Diao, Y.; Zhu, H. An integration of deep learning with feature embedding for protein-protein interaction prediction.
PeerJ 2019, 7, e7126. [CrossRef]
26. Huang, Y.; Wuchty, S.; Zhou, Y.; Zhang, Z. SGPPI: Structure-aware prediction of protein-protein interactions in rigorous conditions
with graph convolutional network. Brief. Bioinform. 2023, 24, bbad020. [CrossRef]
27. Du, X.; Sun, S.; Hu, C.; Yao, Y.; Yan, Y.; Zhang, Y. DeepPPI: Boosting Prediction of Protein-Protein Interactions with Deep Neural
Networks. J. Chem. Inf. Model. 2017, 57, 1499–1510. [CrossRef] [PubMed]
28. Wu, J.; Liu, B.; Zhang, J.; Wang, Z.; Li, J. DL-PPI: A method on prediction of sequenced protein-protein interaction based on deep
learning. BMC Bioinform. 2023, 24, 473. [CrossRef]
29. Zhang, F.; Zhang, Y.; Zhu, X.; Chen, X.; Lu, F.; Zhang, X. DeepSG2PPI: A Protein-Protein Interaction Prediction Method Based on
Deep Learning. IEEE/ACM Trans. Comput. Biol. Bioinform. 2023, 20, 2907–2919. [CrossRef] [PubMed]
30. Ghosh, S.; Mitra, P. MaTPIP: A deep-learning architecture with eXplainable AI for sequence-driven, feature mixed protein-protein
interaction prediction. Comput. Methods Programs Biomed. 2024, 244, 107955. [CrossRef]
31. Soleymani, F.; Paquet, E.; Viktor, H.L.; Michalowski, W.; Spinello, D. ProtInteract: A deep learning framework for predicting
protein-protein interactions. Comput. Struct. Biotechnol. J. 2023, 21, 1324–1348. [CrossRef]
32. Cui, Y.; Dong, Q.; Hong, D.; Wang, X. Predicting protein-ligand binding residues with deep convolutional neural networks. BMC
Bioinform. 2019, 20, 93. [CrossRef]
33. Mylonas, S.K.; Axenopoulos, A.; Daras, P. DeepSurf: A surface-based deep learning approach for the prediction of ligand binding
sites on proteins. Bioinformatics 2021, 37, 1681–1690. [CrossRef]
34. Jendele, L.; Krivak, R.; Skoda, P.; Novotny, M.; Hoksza, D. PrankWeb: A web server for ligand binding site prediction and
visualization. Nucleic Acids Res. 2019, 47, W345–w349. [CrossRef]
35. Kandel, J.; Tayara, H.; Chong, K.T. PUResNet: Prediction of protein-ligand binding sites using deep residual neural network.
J. Cheminform. 2021, 13, 65. [CrossRef] [PubMed]
36. Zhou, Y.; Jiang, Y.; Yang, Y. AGAT-PPIS: A novel protein-protein interaction site predictor based on augmented graph attention
network with initial residual and identity mapping. Brief. Bioinform. 2023, 24, bbad122. [CrossRef] [PubMed]
37. Öztürk, H.; Özgür, A.; Ozkirimli, E. DeepDTA: Deep drug-target binding affinity prediction. Bioinformatics 2018, 34, i821–i829.
[CrossRef] [PubMed]
38. He, T.; Heidemeyer, M.; Ban, F.; Cherkasov, A.; Ester, M. SimBoost: A read-across approach for predicting drug-target binding
affinities using gradient boosting machines. J. Cheminform. 2017, 9, 24. [CrossRef]
39. Ahmed, A.; Mam, B.; Sowdhamini, R. DEELIG: A Deep Learning Approach to Predict Protein-Ligand Binding Affinity. Bioinform.
Biol. Insights 2021, 15, 11779322211030364. [CrossRef]
40. Karlov, D.S.; Sosnin, S.; Fedorov, M.V.; Popov, P. graphDelta: MPNN Scoring Function for the Affinity Prediction of Protein-Ligand
Complexes. ACS Omega 2020, 5, 5150–5159. [CrossRef]
41. Feinberg, E.N.; Sur, D.; Wu, Z.; Husic, B.E.; Mai, H.; Li, Y.; Sun, S.; Yang, J.; Ramsundar, B.; Pande, V.S. PotentialNet for Molecular
Property Prediction. ACS Cent. Sci. 2018, 4, 1520–1530. [CrossRef]
42. Liyaqat, T.; Ahmad, T.; Saxena, C. TeM-DTBA: Time-efficient drug target binding affinity prediction using multiple modalities
with Lasso feature selection. J. Comput. Aided Mol. Des. 2023, 37, 573–584. [CrossRef]
43. Wang, C.; Chen, Y.; Zhang, Y.; Li, K.; Lin, M.; Pan, F.; Wu, W.; Zhang, J. A reinforcement learning approach for protein-ligand
binding pose prediction. BMC Bioinform. 2022, 23, 368. [CrossRef]
44. Nguyen, D.D.; Cang, Z.; Wu, K.; Wang, M.; Cao, Y.; Wei, G.W. Mathematical deep learning for pose and binding affinity prediction
and ranking in D3R Grand Challenges. J. Comput. Aided Mol. Des. 2019, 33, 71–82. [CrossRef]
45. Wang, L.; Zhou, Y.; Chen, Q. AMMVF-DTI: A Novel Model Predicting Drug-Target Interactions Based on Attention Mechanism
and Multi-View Fusion. Int. J. Mol. Sci. 2023, 24, 14142. [CrossRef]
Molecules 2024, 29, 903 17 of 20
46. Popova, M.; Isayev, O.; Tropsha, A. Deep reinforcement learning for de novo drug design. Sci. Adv. 2018, 4, eaap7885. [CrossRef]
[PubMed]
47. Gómez-Bombarelli, R.; Wei, J.N.; Duvenaud, D.; Hernández-Lobato, J.M.; Sánchez-Lengeling, B.; Sheberla, D.; Aguilera-
Iparraguirre, J.; Hirzel, T.D.; Adams, R.P.; Aspuru-Guzik, A. Automatic Chemical Design Using a Data-Driven Continuous
Representation of Molecules. ACS Cent. Sci. 2018, 4, 268–276. [CrossRef] [PubMed]
48. Li, Y.; Zhang, L.; Liu, Z. Multi-objective de novo drug design with conditional graph generative model. J. Cheminform. 2018, 10, 33.
[CrossRef] [PubMed]
49. Born, J.; Manica, M.; Oskooei, A.; Cadow, J.; Markert, G.; Rodríguez Martínez, M. PaccMann(RL): De novo generation of hit-like
anticancer molecules from transcriptomic data via reinforcement learning. iScience 2021, 24, 102269. [CrossRef]
50. Kadurin, A.; Nikolenko, S.; Khrabrov, K.; Aliper, A.; Zhavoronkov, A. druGAN: An Advanced Generative Adversarial Autoen-
coder Model for de Novo Generation of New Molecules with Desired Molecular Properties in Silico. Mol. Pharm. 2017, 14,
3098–3104. [CrossRef]
51. Coley, C.W.; Rogers, L.; Green, W.H.; Jensen, K.F. SCScore: Synthetic Complexity Learned from a Reaction Corpus. J. Chem. Inf.
Model. 2018, 58, 252–261. [CrossRef] [PubMed]
52. Schoenmaker, L.; Béquignon, O.J.M.; Jespers, W.; van Westen, G.J.P. UnCorrupt SMILES: A novel approach to de novo design.
J. Cheminform. 2023, 15, 22. [CrossRef] [PubMed]
53. Wang, X.; Gao, C.; Han, P.; Li, X.; Chen, W.; Rodríguez Patón, A.; Wang, S.; Zheng, P. PETrans: De Novo Drug Design with
Protein-Specific Encoding Based on Transfer Learning. Int. J. Mol. Sci. 2023, 24, 1146. [CrossRef] [PubMed]
54. Monteiro, N.R.C.; Pereira, T.O.; Machado, A.C.D.; Oliveira, J.L.; Abbasi, M.; Arrais, J.P. FSM-DDTR: End-to-end feedback strategy
for multi-objective De Novo drug design using transformers. Comput. Biol. Med. 2023, 164, 107285. [CrossRef] [PubMed]
55. Song, T.; Ren, Y.; Wang, S.; Han, P.; Wang, L.; Li, X.; Rodriguez-Patón, A. DNMG: Deep molecular generative model by fusion of
3D information for de novo drug design. Methods 2023, 211, 10–22. [CrossRef]
56. Macedo, B.; Ribeiro Vaz, I.; Taveira Gomes, T. MedGAN: Optimized generative adversarial network with graph convolutional
networks for novel molecule design. Sci. Rep. 2024, 14, 1212. [CrossRef]
57. Panapitiya, G.; Girard, M.; Hollas, A.; Sepulveda, J.; Murugesan, V.; Wang, W.; Saldanha, E. Evaluation of Deep Learning
Architectures for Aqueous Solubility Prediction. ACS Omega 2022, 7, 15695–15710. [CrossRef]
58. Francoeur, P.G.; Koes, D.R. SolTranNet-A Machine Learning Tool for Fast Aqueous Solubility Prediction. J. Chem. Inf. Model. 2021,
61, 2530–2536. [CrossRef]
59. Zang, Q.; Mansouri, K.; Williams, A.J.; Judson, R.S.; Allen, D.G.; Casey, W.M.; Kleinstreuer, N.C. In Silico Prediction of
Physicochemical Properties of Environmental Chemicals Using Molecular Fingerprints and Machine Learning. J. Chem. Inf. Model.
2017, 57, 36–49. [CrossRef]
60. Tian, H.; Ketkar, R.; Tao, P. ADMETboost: A web server for accurate ADMET prediction. J. Mol. Model. 2022, 28, 408. [CrossRef]
61. Schyman, P.; Liu, R.; Desai, V.; Wallqvist, A. vNN Web Server for ADMET Predictions. Front. Pharmacol. 2017, 8, 889. [CrossRef]
[PubMed]
62. Wei, Y.; Li, S.; Li, Z.; Wan, Z.; Lin, J. Interpretable-ADMET: A web service for ADMET prediction and optimization based on deep
neural representation. Bioinformatics 2022, 38, 2863–2871. [CrossRef] [PubMed]
63. Deng, D.; Chen, X.; Zhang, R.; Lei, Z.; Wang, X.; Zhou, F. XGraphBoost: Extracting Graph Neural Network-Based Features for a
Better Prediction of Molecular Properties. J. Chem. Inf. Model. 2021, 61, 2697–2705. [CrossRef] [PubMed]
64. Mayr, A.; Klambauer, G.; Unterthiner, T.; Hochreiter, S. DeepTox: Toxicity prediction using deep learning. Front. Environ. Sci.
2016, 3, 80. [CrossRef]
65. Li, X.; Xu, Y.; Lai, L.; Pei, J. Prediction of Human Cytochrome P450 Inhibition Using a Multitask Deep Autoencoder Neural
Network. Mol. Pharm. 2018, 15, 4336–4345. [CrossRef]
66. Shaker, B.; Yu, M.S.; Song, J.S.; Ahn, S.; Ryu, J.Y.; Oh, K.S.; Na, D. LightBBB: Computational prediction model of blood-brain-barrier
penetration based on LightGBM. Bioinformatics 2021, 37, 1135–1139. [CrossRef]
67. Tang, Q.; Nie, F.; Zhao, Q.; Chen, W. A merged molecular representation deep learning method for blood-brain barrier permeability
prediction. Brief. Bioinform. 2022, 23, bbac357. [CrossRef]
68. Jang, W.D.; Jang, J.; Song, J.S.; Ahn, S.; Oh, K.S. PredPS: Attention-based graph neural network for predicting stability of
compounds in human plasma. Comput. Struct. Biotechnol. J. 2023, 21, 3532–3539. [CrossRef]
69. Khaouane, A.; Khaouane, L.; Ferhat, S.; Hanini, S. Deep Learning for Drug Development: Using CNNs in MIA-QSAR to Predict
Plasma Protein Binding of Drugs. AAPS PharmSciTech 2023, 24, 232. [CrossRef] [PubMed]
70. Zeng, X.; Zhu, S.; Lu, W.; Liu, Z.; Huang, J.; Zhou, Y.; Fang, J.; Huang, Y.; Guo, H.; Li, L.; et al. Target identification among known
drugs by deep learning from heterogeneous networks. Chem. Sci. 2020, 11, 1775–1797. [CrossRef] [PubMed]
71. Wan, F.; Hong, L.; Xiao, A.; Jiang, T.; Zeng, J. NeoDTI: Neural integration of neighbor information from a heterogeneous network
for discovering new drug-target interactions. Bioinformatics 2019, 35, 104–111. [CrossRef]
72. Luo, Y.; Zhao, X.; Zhou, J.; Yang, J.; Zhang, Y.; Kuang, W.; Peng, J.; Chen, L.; Zeng, J. A network integration approach for
drug-target interaction prediction and computational drug repositioning from heterogeneous information. Nat. Commun. 2017, 8,
573. [CrossRef]
Molecules 2024, 29, 903 18 of 20
73. Luo, H.; Wang, J.; Li, M.; Luo, J.; Peng, X.; Wu, F.X.; Pan, Y. Drug repositioning based on comprehensive similarity measures and
Bi-Random walk algorithm. Bioinformatics 2016, 32, 2664–2671. [CrossRef]
74. Doshi, S.; Chepuri, S.P. A computational approach to drug repurposing using graph neural networks. Comput. Biol. Med. 2022,
150, 105992. [CrossRef] [PubMed]
75. Zeng, X.; Zhu, S.; Liu, X.; Zhou, Y.; Nussinov, R.; Cheng, F. deepDR: A network-based deep learning approach to in silico drug
repositioning. Bioinformatics 2019, 35, 5191–5198. [CrossRef]
76. Jiang, H.J.; Huang, Y.A.; You, Z.H. Predicting Drug-Disease Associations via Using Gaussian Interaction Profile and Kernel-Based
Autoencoder. BioMed Res. Int. 2019, 2019, 2426958. [CrossRef]
77. Ghorbanali, Z.; Zare-Mirakabad, F.; Salehi, N.; Akbari, M.; Masoudi-Nejad, A. DrugRep-HeSiaGraph: When heterogenous
siamese neural network meets knowledge graphs for drug repurposing. BMC Bioinform. 2023, 24, 374. [CrossRef] [PubMed]
78. Suviriyapaisal, N.; Wichadakul, D. iEdgeDTA: Integrated edge information and 1D graph convolutional neural networks for
binding affinity prediction. RSC Adv. 2023, 13, 25218–25228. [CrossRef]
79. Segler, M.H.S.; Preuss, M.; Waller, M.P. Planning chemical syntheses with deep neural networks and symbolic AI. Nature 2018,
555, 604–610. [CrossRef]
80. Liu, B.; Ramsundar, B.; Kawthekar, P.; Shi, J.; Gomes, J.; Luu Nguyen, Q.; Ho, S.; Sloane, J.; Wender, P.; Pande, V. Retrosynthetic
Reaction Prediction Using Neural Sequence-to-Sequence Models. ACS Cent. Sci. 2017, 3, 1103–1113. [CrossRef]
81. Thakkar, A.; Chadimová, V.; Bjerrum, E.J.; Engkvist, O.; Reymond, J.L. Retrosynthetic accessibility score (RAscore)—Rapid
machine learned synthesizability classification from AI driven retrosynthetic planning. Chem. Sci. 2021, 12, 3339–3349. [CrossRef]
82. Wei, J.N.; Duvenaud, D.; Aspuru-Guzik, A. Neural Networks for the Prediction of Organic Chemistry Reactions. ACS Cent. Sci.
2016, 2, 725–732. [CrossRef]
83. Coley, C.W.; Barzilay, R.; Jaakkola, T.S.; Green, W.H.; Jensen, K.F. Prediction of Organic Reaction Outcomes Using Machine
Learning. ACS Cent. Sci. 2017, 3, 434–443. [CrossRef]
84. Gao, H.; Struble, T.J.; Coley, C.W.; Wang, Y.; Green, W.H.; Jensen, K.F. Using Machine Learning To Predict Suitable Conditions for
Organic Reactions. ACS Cent. Sci. 2018, 4, 1465–1476. [CrossRef]
85. Marcou, G.; Aires de Sousa, J.; Latino, D.A.; de Luca, A.; Horvath, D.; Rietsch, V.; Varnek, A. Expert system for predicting reaction
conditions: The Michael reaction case. J. Chem. Inf. Model. 2015, 55, 239–250. [CrossRef]
86. You, Z.H.; Li, S.; Gao, X.; Luo, X.; Ji, Z. Large-scale protein-protein interactions detection by integrating big biosensing data with
computational model. BioMed Res. Int. 2014, 2014, 598129. [CrossRef]
87. Chan, H.C.S.; Shan, H.; Dahoun, T.; Vogel, H.; Yuan, S. Advancing Drug Discovery via Artificial Intelligence. Trends Pharmacol.
Sci. 2019, 40, 592–604. [CrossRef]
88. Muhammed, M.T.; Aki-Yalcin, E. Homology modeling in drug discovery: Overview, current applications, and future perspectives.
Chem. Biol. Drug Des. 2019, 93, 12–20. [CrossRef]
89. Zhang, Y.; Skolnick, J. The protein structure prediction problem could be solved using the current PDB library. Proc. Natl. Acad.
Sci. USA 2005, 102, 1029–1034. [CrossRef]
90. Tang, T.; Zhang, X.; Liu, Y.; Peng, H.; Zheng, B.; Yin, Y.; Zeng, X. Machine learning on protein-protein interaction prediction:
Models, challenges and trends. Brief. Bioinform. 2023, 24, bbad076. [CrossRef]
91. Soleymani, F.; Paquet, E.; Viktor, H.; Michalowski, W.; Spinello, D. Protein-protein interaction prediction with deep learning:
A comprehensive review. Comput. Struct. Biotechnol. J. 2022, 20, 5316–5341. [CrossRef]
92. Li, S.; Wu, S.; Wang, L.; Li, F.; Jiang, H.; Bai, F. Recent advances in predicting protein-protein interactions with the aid of artificial
intelligence algorithms. Curr. Opin. Struct. Biol. 2022, 73, 102344. [CrossRef]
93. Tripathi, N.; Goshisht, M.K.; Sahu, S.K.; Arora, C. Applications of artificial intelligence to drug design and discovery in the big
data era: A comprehensive review. Mol. Divers. 2021, 25, 1643–1664. [CrossRef]
94. Dhakal, A.; McKay, C.; Tanner, J.J.; Cheng, J. Artificial intelligence in the prediction of protein-ligand interactions: Recent advances
and future directions. Brief. Bioinform. 2022, 23, bbab476. [CrossRef]
95. Nicolaou, C.A.; Kannas, C.; Loizidou, E. Multi-objective optimization methods in de novo drug design. Mini Rev. Med. Chem.
2012, 12, 979–987. [CrossRef]
96. Zhong, F.; Xing, J.; Li, X.; Liu, X.; Fu, Z.; Xiong, Z.; Lu, D.; Wu, X.; Zhao, J.; Tan, X.; et al. Artificial intelligence in drug design. Sci.
China. Life Sci. 2018, 61, 1191–1204. [CrossRef]
97. Hessler, G.; Baringhaus, K.H. Artificial Intelligence in Drug Design. Molecules 2018, 23, 2520. [CrossRef]
98. Schneider, G.; Funatsu, K.; Okuno, Y.; Winkler, D. De novo Drug Design—Ye olde Scoring Problem Revisited. Mol. Inform. 2017,
36, 1681031. [CrossRef]
99. Wang, L.; Ding, J.; Pan, L.; Cao, D.; Jiang, H.; Ding, X. Artificial intelligence facilitates drug design in the big data era. Chemom.
Intell. Lab. Syst. 2019, 194, 103850. [CrossRef]
100. Yang, X.; Wang, Y.; Byrne, R.; Schneider, G.; Yang, S. Concepts of Artificial Intelligence for Computer-Assisted Drug Discovery.
Chem. Rev. 2019, 119, 10520–10594. [CrossRef]
101. Yu, E.; Xu, Y.; Shi, Y.; Yu, Q.; Liu, J.; Xu, L. Discovery of novel natural compound inhibitors targeting estrogen receptor α by an
integrated virtual screening strategy. J. Mol. Model. 2019, 25, 278. [CrossRef]
Molecules 2024, 29, 903 19 of 20
102. Zhong, W.; Zhao, L.; Yang, Z.; Yu-Chian Chen, C. Graph convolutional network approach to investigate potential selective Limk1
inhibitors. J. Mol. Graph. Model. 2021, 107, 107965. [CrossRef]
103. Zhou, Y.; Wang, F.; Tang, J.; Nussinov, R.; Cheng, F. Artificial intelligence in COVID-19 drug repurposing. Lancet Digit. Health
2020, 2, e667–e676. [CrossRef]
104. Pan, X.; Lin, X.; Cao, D.; Zeng, X.; Yu, P.S.; He, L.; Nussinov, R.; Cheng, F. Deep learning for drug repurposing: Methods, databases,
and applications. Wiley Interdiscip. Rev. Comput. Mol. Sci. 2022, 12, e1597. [CrossRef]
105. Dong, J.; Zhao, M.; Liu, Y.; Su, Y.; Zeng, X. Deep learning in retrosynthesis planning: Datasets, models and tools. Brief. Bioinform.
2022, 23, bbab391. [CrossRef] [PubMed]
106. Lee, A.A.; Yang, Q.; Sresht, V.; Bolgar, P.; Hou, X.; Klug-McLeod, J.L.; Butler, C.R. Molecular Transformer unifies reaction
prediction and retrosynthesis across pharma chemical space. Chem. Commun. 2019, 55, 12152–12155. [CrossRef]
107. Yan, X.; Liu, Y. Graph-sequence attention and transformer for predicting drug-target affinity. RSC Adv. 2022, 12, 29525–29534.
[CrossRef]
108. Lee, M. Recent Advances in Deep Learning for Protein-Protein Interaction Analysis: A Comprehensive Review. Molecules 2023,
28, 5169. [CrossRef]
109. Lin, P.; Yan, Y.; Huang, S.Y. DeepHomo2.0: Improved protein-protein contact prediction of homodimers by transformer-enhanced
deep learning. Brief. Bioinform. 2023, 24, bbac499. [CrossRef] [PubMed]
110. Kang, Y.; Elofsson, A.; Jiang, Y.; Huang, W.; Yu, M.; Li, Z. AFTGAN: Prediction of multi-type PPI based on attention free
transformer and graph attention network. Bioinformatics 2023, 39, btad052. [CrossRef]
111. Zhang, P.; Wei, Z.; Che, C.; Jin, B. DeepMGT-DTI: Transformer network incorporating multilayer graph information for Drug-
Target interaction prediction. Comput. Biol. Med. 2022, 142, 105214. [CrossRef]
112. Qian, H.; Lin, C.; Zhao, D.; Tu, S.; Xu, L. AlphaDrug: Protein target specific de novo molecular generation. PNAS Nexus 2022,
1, pgac227. [CrossRef]
113. Radford, A.; Narasimhan, K.; Salimans, T.; Sutskever, I. Improving Language Understanding by Generative Pre-Training. 2018.
Available online: https://www.mikecaptain.com/resources/pdf/GPT-1.pdf (accessed on 14 January 2024).
114. Wang, Y.; Zhao, H.; Sciabola, S.; Wang, W. cMolGPT: A Conditional Generative Pre-Trained Transformer for Target-Specific De
Novo Molecular Generation. Molecules 2023, 28, 4430. [CrossRef]
115. Chithrananda, S.; Grand, G.; Ramsundar, B. ChemBERTa: Large-scale self-supervised pretraining for molecular property
prediction. arXiv 2020, arXiv:2010.09885.
116. Devlin, J.; Chang, M.-W.; Lee, K.; Toutanova, K. Bert: Pre-training of deep bidirectional transformers for language understanding.
arXiv 2018, arXiv:1810.04805.
117. Liu, Z.; Roberts, R.A.; Lal-Nag, M.; Chen, X.; Huang, R.; Tong, W. AI-based language models powering drug discovery and
development. Drug Discov. Today 2021, 26, 2593–2607. [CrossRef] [PubMed]
118. Wu, Z.; Jiang, D.; Wang, J.; Zhang, X.; Du, H.; Pan, L.; Hsieh, C.Y.; Cao, D.; Hou, T. Knowledge-based BERT: A method to extract
molecular features like computational chemists. Brief. Bioinform. 2022, 23, bbac131. [CrossRef]
119. Wang, S.; Guo, Y.; Wang, Y.; Sun, H.; Huang, J. Smiles-bert: Large scale unsupervised pre-training for molecular property
prediction. In Proceedings of the 10th ACM International Conference on Bioinformatics, Computational Biology and Health
Informatics, Niagara Falls, NY, USA, 7–10 September 2019; pp. 429–436.
120. Schwaller, P.; Laino, T.; Gaudin, T.; Bolgar, P.; Hunter, C.A.; Bekas, C.; Lee, A.A. Molecular Transformer: A Model for Uncertainty-
Calibrated Chemical Reaction Prediction. ACS Cent. Sci. 2019, 5, 1572–1583. [CrossRef]
121. Andronov, M.; Voinarovska, V.; Andronova, N.; Wand, M.; Clevert, D.-A.; Schmidhuber, J. Reagent prediction with a molecular
transformer improves reaction data quality. Chem. Sci. 2023, 14, 3235–3246. [CrossRef]
122. Schwaller, P.; Petraglia, R.; Zullo, V.; Nair, V.H.; Haeuselmann, R.A.; Pisoni, R.; Bekas, C.; Iuliano, A.; Laino, T. Predicting
retrosynthetic pathways using transformer-based models and a hyper-graph exploration strategy. Chem. Sci. 2020, 11, 3316–3325.
[CrossRef]
123. Mamoshina, P.; Vieira, A.; Putin, E.; Zhavoronkov, A. Applications of Deep Learning in Biomedicine. Mol. Pharm. 2016, 13,
1445–1454. [CrossRef]
124. Lu, J.; Wang, C.; Zhang, Y. Predicting Molecular Energy Using Force-Field Optimized Geometries and Atomic Vector Representa-
tions Learned from an Improved Deep Tensor Neural Network. J. Chem. Theory Comput. 2019, 15, 4113–4121. [CrossRef]
125. Cai, C.; Wang, S.; Xu, Y.; Zhang, W.; Tang, K.; Ouyang, Q.; Lai, L.; Pei, J. Transfer Learning for Drug Discovery. J. Med. Chem. 2020,
63, 8683–8694. [CrossRef]
126. Altae-Tran, H.; Ramsundar, B.; Pappu, A.S.; Pande, V. Low Data Drug Discovery with One-Shot Learning. ACS Cent. Sci. 2017, 3,
283–293. [CrossRef]
127. Krizhevsky, A.; Sutskever, I.; Hinton, G.E. ImageNet classification with deep convolutional neural networks. Commun. ACM 2017,
60, 84–90. [CrossRef]
128. Chang, M. AI for Drug Development and Well-Being. 2020. Available online: http://ctrisoft.net/StatisticiansOrg/AI/
AIforWellbingebook5.5x8.5in.pdf (accessed on 14 January 2024).
129. Erhan, D.; Bengio, Y.; Courville, A.C.; Vincent, P. Visualizing Higher-Layer Features of a Deep Network; University of Montreal:
Montreal, QC, USA, 2009.
Molecules 2024, 29, 903 20 of 20
130. Ribeiro, M.T.; Singh, S.; Guestrin, C. “Why Should I Trust You?”: Explaining the Predictions of Any Classifier. In Proceedings of
the 22nd ACM SIGKDD International Conference on Knowledge Discovery and Data Mining, San Francisco, CA, USA, 13–17
August 2016; pp. 1135–1144.
131. Lundberg, S.M.; Lee, S.-I. A Unified Approach to Interpreting Model Predictions. In Proceedings of the Neural Information
Processing Systems, Long Beach, CA, USA, 4–9 December 2017.
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.