You are on page 1of 27

Briefings in Bioinformatics, 2023, 24(3), 1–27

https://doi.org/10.1093/bib/bbad120
Advance access publication date 6 April 2023
Review

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Machine learning for synergistic network pharmacology:
a comprehensive overview
Fatima Noor† , Muhammad Asif† , Usman Ali Ashfaq, Muhammad Qasim and Muhammad Tahir ul Qamar
Corresponding author. Muhammad T. u. Qamar, Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan.
E-mail: tahirulqamar@gcuf.edu.pk
† F. Noor and M. Asif contributed equally.

Abstract
Network pharmacology is an emerging area of systematic drug research that attempts to understand drug actions and interactions
with multiple targets. Network pharmacology has changed the paradigm from ‘one-target one-drug’ to highly potent ‘multi-target
drug’. Despite that, this synergistic approach is currently facing many challenges particularly mining effective information such as drug
targets, mechanism of action, and drug and organism interaction from massive, heterogeneous data. To overcome bottlenecks in multi-
target drug discovery, computational algorithms are highly welcomed by scientific community. Machine learning (ML) and especially
its subfield deep learning (DL) have seen impressive advances. Techniques developed within these fields are now able to analyze and
learn from huge amounts of data in disparate formats. In terms of network pharmacology, ML can improve discovery and decision
making from big data. Opportunities to apply ML occur in all stages of network pharmacology research. Examples include screening
of biologically active small molecules, target identification, metabolic pathways identification, protein–protein interaction network
analysis, hub gene analysis and finding binding affinity between compounds and target proteins. This review summarizes the premier
algorithmic concepts of ML in network pharmacology and forecasts future opportunities, potential applications as well as several
remaining challenges of implementing ML in network pharmacology. To our knowledge, this study provides the first comprehensive
assessment of ML approaches in network pharmacology, and we hope that it encourages additional efforts toward the development
and acceptance of network pharmacology in the pharmaceutical industry.

Keywords: network pharmacology, artificial intelligence, machine learning, deep learning, drug discovery

BACKGROUND [4–6]. Multi-target drugs have gained considerable interest in the


Drug discovery and development together is the whole pro- last decade because of their potential to treat diseases involving
cess of identifying novel and effective drug candidates and interaction of multiple genes, thus yielding successful outcome
considered as a key part of clinical pharmacology [1]. Drug as compared to single-target drugs [7].
discovery involves many different processes including screening Network graphs provide a convenient conceptual framework
of chemical libraries and identification of biologically active for systems-level modeling, integrating and mining of high-
small molecules [2]. On the other hand, drug development throughput experimental datasets. This helps in understanding
includes optimization of small molecules, toxicological studies and gaining insights into different types of molecular relationship,
on different microorganisms and animals, clinical trials and such as how genes are linked to various diseases or interactions
ultimately regulatory approval [3]. Over the last decade, the between drugs and their cellular targets [8]. Systems or network
‘one disease–one target–one drug’ paradigm has dominated the biology has proven useful for deciphering fundamental research
drug discovery approaches. However, this paradigm has several questions, such as how perturbations in the cellular networks
limitations and recent advancements in systems biology has lead to certain phenotypes, including human diseases [9, 10].
shifted the focus from ‘single-drug target’ to ‘multi-target drugs’ In network context, the efficacy of multi-target therapy can be

Fatima Noor is currently an MS Bioinformatics scholar at the Integrative OMICS & Molecular Modeling Lab, Department of Bioinformatics and Biotechnology,
Government College University Faisalabad, Pakistan. Her current research focuses on the development of machine learning-based tools for integrated biomedical
data analysis.
Muhammad Asif is an Assistant Professor at the Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Pakistan. He has
PhD in Systems Biology and Bioinformatics. He is leading a biomedical data analysis lab, where he is mainly focused on developing machine and deep learning
methods/software for biomedical data integration and analysis, in particular for bulk sequencing, and single-cell omics.
Usman Ali Ashfaq is a Professor at the Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Pakistan. He is leading a
health biotechnology lab, where he is mainly focused on the diagnosis and treatment of in fectious and metabolic diseases using advanced Bioinformatics
approaches.
Muhammad Qasim is a Professor and currently serving as the Chairperson of the Department of Bioinformatics and Biotechnology, Government College
University Faisalabad, Pakistan. His research interests include high-throughput genome sequencing data analysis, genomic medicine, and integrated
Bioinformatics.
Muhammad Tahir ul Qamar is an Assistant Professor and leading Integrative OMICS & Molecular Modeling Lab at the Department of Bioinformatics and
Biotechnology, Government College University Faisalabad, Pakistan. He received his Ph.D. degree in Bioinformatics from Huazhong Agricultural University, Wuhan,
China. His research focuses on developing methods, resources, and software for bulk/single-cell sequencing data analysis, with a particular focus on
Pan-genomics.
Received: August 2, 2022. Revised: March 5, 2023. Accepted: March 10, 2023
© The Author(s) 2023. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com
2 | Noor et al.

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Figure 1. Workf low representing the network pharmacology research to decipher the biologically active molecules for the treatment of incurable
diseases. At first, it analyzes the targets from different resources including experimental data, public data available in databases, and then performs
network analysis. Based on integrative analysis, it identified hub genes specifically involves in the dysregulation of pathway that leads to disease
condition. Finally, it performs experimental and computational validations.

understood from a robustness of disease networks to deal with when their combined effect is greater than their individual effect
single node perturbations, due to inherent diversity and redun- [17]. Synergistic activity may emerge from a set of targeted
dancy of compensatory signaling pathways that result in highly proteins, related mechanisms, or compounds combinations.
resilient network architecture with modular and interconnected In network pharmacology, synergistic action is due to the
topology. Therefore, network-based drug discovery seeks for drug combinatorial effects of active compounds on multiple targets
target combinations to perturb a specific subset of nodes in the identified by network topological features [18, 19]. Network
disease-associated networks to inhibit the bypass mechanisms pharmacology aims to understand ‘network-target, multiple-
at systems level [11]. These recent developments have resulted component-therapeutics’ by ‘compound-protein/gene-disease’
in moving away from the traditional ‘one target’ strategy toward network revealing the regulation principles of small molecules
network pharmacology [12]. in a high-throughput manner and analysis of drug combinations,
Network pharmacology is recent frontier in systematic drug hence synergy.
research as it tends to elucidate the drug actions as well as Identification of disease and compound-related target is a
their interaction with multiple targets [13]. This approach is preliminary step to figure out the anti-disease-related compound
combining, experimental, computational and clinical advances targets [20, 21]. Following that, the protein–protein interaction
to increase the clinical efficacy of drugs [14]. It has now (PPI) network of target proteins is constructed, analyzed and
been dubbed as ‘next paradigm in drug development’ as it visualized to get a better understanding of the proper function-
presents a comprehensive grasp of systems biology with network ing of molecular mechanisms perturbed in disease states [21].
theories [15]. This systematic approach revolves around the Later, it extracted hub genes on the basis of their degree of
identification of synergistic and mechanistically related co- connective from gene networks. Finally, further network valida-
targets of active compounds in a network perspective (Figure 1) tion is carried out to evaluate the interaction between active
[16]. Synergistic is a multi-objective approach where synergy compounds and their potential disease targets [22, 23]. In sum-
can be observed from a network pharmacology perspective. mary, network pharmacology can assist in evaluating the effi-
Synergistic describes the interaction of two or more compounds ciency of multi-component, multi-target compound formulas and
Machine learning for synergistic network pharmacology | 3

exploring more therapeutic strategies by targeting a specific net- data resources that can provide a helping hand in this endeavor
work. Despite that, this approach is facing challenges including [39]. However, they need advance approaches to smartly process
limited technological advancements and knowledge to identify this available data. The advancements in ML have made big data
novel drug targets, lack of synergistic drugs and poor quality of analytics significantly must easier, as there is a plethora of ML
databases in terms of data management [24, 25]. To bridge the gap, approaches that may assist in extracting meaningful features,

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
computational algorithms must be exploited to overcome the patterns and structures from large biomedical datasets [40]. Guo
limitations of network pharmacology. et al. [41] proposed an unsupervised ML approach to explore
Machine learning (ML) approaches are becoming popular the cellular functional similarity of XiaoErFuPi granules. They
across all facets of sciences. Fundamentally, ML is the practice of used hierarchical clustering and then classify the compounds
using algorithms to parse data, learn from it and then accordingly into several different modules with similar therapeutic functions.
make a prediction about the future state of any new dataset [26, Later, the predicted modules have similar function fingerprints
27]. In ML data and algorithms like K-Nearest Neighbors (kNN), with various FDA-approved drugs. Thus, the prediction of small
Decision Tree, Support Vector Machine (SVM), Naïve Bayes and molecules based on the similar function fingerprints proven more
Hidden Markov Model (HMM) are used to imitate the way how effective as compared to applying simple criteria of drug likeness
humans learn [28, 29]. Advancements in ML has paved the road and bioavailability. Their study yielded successful outcome by
to the discovery of synergistic drugs [30]. Big data and cutting- providing a new insight to the synergistic activity of XiaoErFuPi
edge algorithms are now readily available, which has increased granules against dyspepsia.
interest and led to substantial advancements in the application of Synergistic combinations of drugs can overcome toxicity
ML in network pharmacology [31]. All pharmaceutical companies and other side effects associated with high doses of single
are routinely combining multiple ML algorithms to streamline drugs, by either countering biological compensation, sparing
the process of discovering new drugs. ML approaches can foster doses on each compound, or accessing context-specific multi-
data-driven decision-making and has the potential to accelerate target mechanisms [42]. The growing availability of large-
the process and decrease the error rates in the drug discovery scale, high-throughput data has prompted the development
process [32, 33]. Ever since its conception, ML has undergone a of computational approaches that can effectively predict
half-century of development and has been effectively applied to the potential synergistic drug combinations. Considering the
a wide range of industrial and technological domains. limitations of previous computational methods, Li et al. [43]
ML techniques are increasingly being implemented to deal developed a new model named Siamese Network and Random
with the limitations of network pharmacology process owing Matrix Projection for Anti-Cancer Drug Combination prediction
to advancements, globalization and the adaptation of big data (SNRMPACDC). Besides, SNRMPACDC achieved the Area under the
available for producing meaningful insights. The existing reviews ROC Curve (AUC) of 0.91 ± 0.03 and the Area Under the Precision–
are focused merely on drug–target or drug–drug interaction pre- Recall curve (AUPR) of 0.62 ± 0.05 in 5-fold cross-validation of
diction. The significant research on finding drug interaction with classification prediction of synergistic or not. These results
a network of targets (instead of single target) is missing. Currently, indicated that SNRMPACDC would be an effective approach
to the best of our knowledge we do not have any tool or classifier to infer potential anticancer synergistic drug combinations.
that could predict the interaction of a drug compound with a Similarly, Chen et al. [44] developed a novel algorithm named
network of targets. The main objective of this review is to cover the as Network-based Laplacian regularized Least Square Synergistic
basics of ML methods for network pharmacology that may stim- drug combination prediction (NLLSS) for prediction of potential
ulate new research trends. Our review discusses the application synergistic drug combinations by integrating different kinds of
of ML as a benchmark to advance the multi-target drug discovery information including drug–target interactions, drug chemical
by modernizing network pharmacology research. We also address structures and known synergistic drug combinations. Compared
crosstalk between different databases/tools used in ML to improve with the traditional method of splicing drug features into drug
decision-making and deducing risk failures in multi-target drug combination features, these computational models can improve
discovery, along with the future of ML approaches in network the interpretability of drug combination features to a certain
pharmacology research. Brief ly, this review article intends to open extent.
new avenues for the discovery of putative treatment options by Nowadays, gene expression analysis is frequently used to cap-
combining powerful ML techniques with network pharmacology. ture the pathophysiological mechanisms of diseases as well as to
uncover genes involved in the disease conditions. The technolo-
gies of microarray and RNA-seq has produced a vast quantity of
REVOLUTIONIZING THE NETWORK gene expression data for a variety of diseases. A huge quantity
PHARMACOLOGY: BIG DATA AND ML of gene expression data has been produced with the emergence
Big data is a collection of datasets which are big and complex of NGS technologies and stored in publicly available databases
to be evaluated by using traditional data analysis software, tools such as The Cancer Genome Atlas (TCGA) [45], ArrayExpress [46]
and procedures [34, 35]. Volume, velocity and variety are the three and Gene Expression Omnibus (GEO) [47]. Voluminous data are
fundamental characteristics of big data, wherein volume ref lects produced from these platforms. Regarding this, ML techniques are
the massive amount and quantity of data generated, velocity warmly welcomed by the scientific community to identify unique
describes the rate at which these data are multiplied, and variety patterns that are relevant to a specific biological problem. Recent
signifies the heterogeneity found in the huge datasets [36]. Day studies imply kNN algorithm on gene expression data for the iden-
after day, a wealth of biological data is generated by virtue of tification of prognostic genes underlying the disease condition
high throughput technologies such as sequencing technologies, [48]. However, unfortunately, we do not have any successful stud-
contributing to the transformation of modern drug development ies in network pharmacology, where ML assisted gene expression
through network pharmacology into the big data era [37, 38]. Tar- data is being used for the identification of therapeutic targets. Sev-
get prediction is the preliminary step in network pharmacology eral functionally significant pathways involved in a particular dis-
pipelines. Now researchers have access to plenty of biomedical ease condition are unknown. This information is now expanded
4 | Noor et al.

due to an unprecedented increased in high-throughput data. ML and quantity of data determine the effectiveness of final predic-
approaches are widely used by researchers to identify specific tive model. Following that, it generates different types of robust
pathways because ML is capable of handling big and complex data mathematical descriptors to predict the toxicity and bioactivity
sets. Baranwal et al. [49] applied deep learning (DL) algorithms of molecules from their structure. For analysis, the model is
for metabolic pathways prediction. They used graph Convolution trained with training datasets to analyze how it respond to data

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Neural Network (CNN) with RF to predict the set of pathways which it evaluate/seen before. Lastly, model testing is carried out
to which a specific small molecule may belong. At present, the to validate the raw data and to evaluate the performance of
applications of DL and cloud computing sparked a lot of interest predictive model [56, 58]. Altogether, this workf low provides the
in network pharmacology, notably in screening bioactive com- base for multi-target drug discovery.
pounds, targets and action pathways of single drugs. Several ML is less obsessed with hypothetical breakthroughs and more
studies employed ML or DL to predict either drug–drug or drug– focused on translating medical data into studies that can be
target interactions. For example, Monteiro et al. [50] employed reused. Nowadays, countless ML approaches are used in network
CNN to extract a 1D representation from 1D raw data, amino acid pharmacology to analyze the drug action (Figure 3), including
sequence and SMILES. The extracted features representing the Autoencoder (AE), HMM, Multi-Layer Perceptron (MLP), RF, SVM,
local dependencies were fed to a fully connected neural network. kNN and more important DL to improve decision making in net-
The proposed DL architecture model resulted in a high sensitivity work pharmacology. AE is a subtype of Artificial Neural Network
(0.861), specificity (0.961) and overall accuracy of 0.923 when com- (ANN) where neural network is trained to reconstruct the input
pared with the other traditional models. Heba et al. [51] proposed in an unsupervised manner. AE is highly capable of spotting
an ML approach that exploits the both sequence and protein fundamental structure of input data rather than memorizing
structural features to predict drug–target interactions. Authors them [59, 60]. AE has a key role to play in the discovery of small
evaluated the performance of SVM, RF, ensemble and DL tech- molecule as drug candidate against incurable diseases. Recently,
niques in predicting the drug target interaction. Heba et al. found Hu et al. [61] used fully connected neural networks to construct
that ensemble learning methods (Light-Boost and ExtraTree) out- drug-likeness classification models with deep AE to initialize
performed the previously reportedly methods with an accuracy model parameters. Their study provided a 91.04% classification
of 98% and 0.97 F1-score, which is 2% higher average accuracy accuracy of drug-like/nondrug–like models 91.04% on World Drug
than the existing methods. Recently, Zong et al. [52] have also pre- Index (WDI)/Available Chemicals Directory (ACD) databases while
sented a comprehensive benchmark, called BETA for drug–target 91.20% classification accuracy of drug-like/nondrug–like models
prediction. A large multipartite network, along with different on MACCS-II Drug Data Report (MDDR)-ZINC database. Following
validation techniques were employed in BETA, which may help in that, Emdadi et al. [62] introduced Auto-HMM-LMF, a novel and an
deciding the computational strategies for drug–target interaction efficient feature-selection method used by researchers to predict
prediction. Network-based methods along with structure and the response of specific drug. Auto-HMM-LMF used HMM and AE
sequence-based methods were used for the purpose of evaluating for feature selection. HMM is a general modeling technique used
proposed benchmark based on the input data. Overall, the results in unsupervised learning for the modeling of biological sequences
demonstrated that in general, the best methods for network- [63]. A combination of HMM with AE assists the researcher to
based and structure- and sequence-based methods are DeepPur- analyze the properties of active compounds. However, no study
pose (CNN) [53] (average AUCROC: 88.01%, PRAUC: 84.44%, preci- to date used AE and HMM for multi-target drug discovery in a
sion: 73.40%, recall: 86.73%, F1: 79.49%, general score: 83.98%) and network pharmacology perspective. Therefore, the prediction of
NeoDTI [54] (average AUCROC: 86.52%, PRAUC: 83.32%, precision: drug-likeness properties of compounds and action mechanism of
71.40%, recall: 80.78%, F1: 74.19%). It, in a sense, represented an active compounds using AE and HMM might help in the screening
indirect connection between topology-, structure- and sequence- of potential active compounds for further analysis. A few common
based features for drugs and proteins in drug development con- models, including as SVM, RF and DL, are described in detail in
texts. order to promote multi-target drug discovery by combining ML
Despite these efforts, new tools with ability to predict drug approaches with network pharmacology.
interaction with a network of targets are required. In this review,
along with network pharmacology we have discussed highly Support Vector Machines
sophisticated ML and DL methods. The main objective was to SVMs are supervised learning methods and can be used for both
stress on surging interests in network pharmacology that may classification and regression analysis [64, 65]. SVM creates a n-
stimulate new research trends. dimensional feature space, where n is the number of features.
SVM aims for finding a hyperplane that can classify data into
distinct classes with a minimal error (Figure 4) [66]. The goal of
ML METHODS FOR IMPROVING SVM is to divide the datasets into classes to find a maximum
MULTI-TARGET DRUG DISCOVERY IN marginal hyperplane. First, SVM will generate hyperplanes iter-
NETWORK PHARMACOLOGY atively that segregates the classes in best way. Then, it will choose
The advent of ML techniques was first unveiled in 1950 and has the hyperplane that separates the classes optimally [67]. SVM can
already been employed in modern fields including bioinformatics, be liner and non-liner, which can be defined by choosing a kernel.
systems biology, network pharmacology and many others [55]. In SVM, X represented the feature vector, or the input provided
Particularly, ML approaches can be applied in five main steps for to SVM, whereas Y represents the output or class, i.e. Y ∈ {−1, 1}.
multi-target drug discovery (Figure 2) including (i) data collection, At the same moment, X ∈ Rn , where n is a dimension feature
(ii) retrieving different types of molecular descriptors, (iii) figure vector. The subject of classifying binary class is explored here.
out optimal subset of variables, (iv) Training of model and (v) Parameters in SVM w and b have considered for learning data
validation of trained model [56, 57]. At first, it collects infor- in training set. w is normal to the line and known as the weight
mation about small biologically active molecules from different vector, whereas b is the bias. The main goal of the learning data
resources. Data collection is extremely important. The quality is to find the optimal set of weights, which can ideally produce
Machine learning for synergistic network pharmacology | 5

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Figure 2. Workf low of ML methodology to improve the multi-target drug discovery process. At first, it collected data and then generate mathematical
descriptors to place corresponding patches close to each other. Later, it trained the model and then cross-validate the model.

the correct output for the relative input. Xi and are the ith sam- Similarly,
ple in the dataset, whereas the f (X) can also be represented as
follows: 2  
minw = Yi wT Xi + b ≥ 1 for i = 1, 2, 3, .., N (3.4)
 ||w||
≥ 0 Yi = 1
f (Xi ) = (3.1)
< 0 Yi = −1
It then generates a quadratic optimization model, such as
  minw
 
The output can be described as f (Xi ) Y(i) + b ≥ 1. Obtain a w,b2
.therebyY(i) wT Xi + b ≥ 1, for i = 1, 2, 3, .., N.
weight vector w such that the discriminant function is satisfied Because the above-mentioned equation was indeed a hard
with the linearly separable data Xi labeled into two classesYi = margin SVM, we can overcome this issue by using the linearly
{−1, 1}: separable technique. Simply use slack variables as constraints.
Each sample in the training data has its own slack variable.
f (Xi ) = wT Xi + b (3.2) Therefore,

min ||w|| N
+C (3.5)
It separates the categories for i = 1, 2, 3, ..N. The distance w, b2 ∈i
i
between two hyperplanes wT X + b = −1 and wT X + b = 1should  T 
maximized the separation between two endpoints. As a result, Yi w Xi + b ≥ 1 − ∈i for i = 1 2, 3, . . . , N (3.6)
2
Distance = maxw ||w|| 2
. Later, it solves the maxw ||w|| minw ||w||
2
:
where C is a regularization parameter. C parameter adds a penalty
2 ≥1 if Y =+1 for each misclassified data point. For large values of C, the
maxw = wT Xi + b≤−1 if iY =−1 for i = 1, 2, 3..N (3.3)
||w|| i
optimization will choose a smaller-margin hyperplane if that
6 | Noor et al.

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023

Figure 3. Overview of ML tools and techniques for multi-target drug discovery. Unsupervised techniques are used to construct models that enable data
clustering, whereas supervised learning techniques including regression and classifier approaches are used to solve queries that demand the prediction
of data categories or continuous variables. Thus, both provide a helping hand in network pharmacology research.

hyperplane does a better job of getting all the training points The SVM, with its kernels, is frequently employed in net-
classified correctly. Conversely, a very small value of C will work pharmacology to identify effective compounds against com-
cause the optimizer to look for a larger-margin separating plex diseases. Dai et al. [68] used network pharmacology-based
hyperplane, even if that hyperplane misclassifies more points. approach for the identification of traditional Chinese medicinal
Adding a function ∅ in order to provide additional mapping formula against Huntington’s disease. Later, they used SVM to
f lexibility. Consequently, it translates diverse characteristics validate their findings. To construct well-trained Quantitative
such as original space to high-dimensional space. The updated Structure–Activity Relationship (QSAR) models they applied SVM
equation is algorithms on generated descriptors. Their study combines ML-
algorithms with network pharmacology which lays a new sci-
 
Thus, Yi wT φ Xi + b ≥ 1 − ∈i for i = 1, 2, 3, . . . , N (3.7) entific foundation to analyze the efficacy of small molecules
Machine learning for synergistic network pharmacology | 7

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Figure 4. Operational workf low of SVM.

for exploring more multi-target drugs in future. Furthermore, consists of M = M1 , M2 , . . . , Mn , N = N1 , N2 , . . . , Nn :


Zernov et al. [69] used SVM to predict the drug-likeness of small
molecules. It is worth noting that SVM model used for prediction ⎡ ⎤
M1 N1
of drug-likeness property of small molecules is much better than
⎢ M2 N2 ⎥
using traditional QSAR. Lei et al. [70] used SVM to predict the oral ⎢ ⎥
⎢ ⎥
⎢ M3 N3 ⎥
acute toxicity in mammals. However, currently in network phar- P= ⎢ ⎥ (3.8)
⎢ . . ⎥
macology no study to date implementing SVM algorithm to access ⎢ ⎥
⎣ . . ⎦
the drug-likeness and toxicity of small molecules. Therefore, SVM
Mn Nn
algorithms can be used in network pharmacology to predict the
efficacy of active compounds.

where M1 represents the feature of first sample, N1 represents


the features of second sample, whereas n represents the infinite
Random Forest
features. Thus, the main objective is to create an RF by classifying
Random Forest (RF) is a powerful and versatile supervised
the sample set i.e., P. Following the sample set P, a list of subsets
ML algorithm that grows and combines multiple decorrelated
is created with random values.
decision trees to create a ‘forest’. It uses bagging and feature
Alternate samples with n trained examples from M, N then Ma ,
randomness when building each individual tree to try to create
Na . To make a prediction at a new point i.e.,  o to bth RF tree:
an uncorrelated forest of trees whose prediction is more accurate
than that of any individual tree [71]. Over-fitting can emerge in
regression and classification tasks, which may consequent into 1 B
f̂ = fb (o) (3.9)
a biased classifier. The chances of over-fitting may be reduced B b=1

for some datasets by applying RF algorithm. RF revolves around


the implementation of trained algorithms approaches such as
bagging [72]. The main theme of bagging method is that the A simple majority can be used in classification trees. Lastly,
combination of learning models improves the final output. because there is no more incremental bias in the RF model, it
Generally, the datasets consist of a matrix of trained samples generates superior results and lowers variation in the model.
for the construction of a classification model. The trained sample Therefore, the standard deviation form of the equation for an
8 | Noor et al.

deep architecture and training it; (v) the performance of traiend


model is evaluated; and, lastly (vi), deployment of the model. DL
algorithms has recently been employed in a variety of scientific
domains. DL has a neural network architecture that comprises a
set of layers and allows input to be processed between them [79].

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
DL models make use of many different algorithms. Each algorithm
has its own pros and cons and is well suited for a specialized task.
Therefore, it is necessary to develop a thorough understanding
of all DL algorithms for achieving better results. MLPs, Convolu-
tional Neural Networks (CNNs), Deep Neural Networks (DNNs)
and Recurrent Neural Networks (RNNs) are extensively used by
Figure 5. The RF constructed a visual decision tree to retrieve calculations
for each sample and then vote on the best outcomes. It first test the sam-
researchers for drug discovery which are described below.
ple input and then average all predictions. Based on the voting/averaging The MLP model is a fully connected class of feedforward
final prediction is made [Figure is adopted/modified from Dara et Al. [72] ANN. ANN is an interconnected group of nodes that mimics
© (2021) Springer]. the way nerve cells work in the human brain. MLP generates
output depending on a set of input data [80]. MLP composed
of interconnected neurons transferring information from one
independent regression tree on x is neuron/layer to another just like in brain. A single neuron in this
network is called perceptron. Each perceptron in MLP accepted n
B  2 features from input layers (a = a1 , a2 , a3 , . . . an ). All these features
 b=1 fb (o) − f̂
Standard deviation = (3.10) are interconnected with some specific weights. The input features
B−1 are then passed to a new non-linear function (z) which mainly
aims to predict the weights for all input features simultaneously:
where B is an arbitrary parameter. The RF algorithm integrates
many different decision trees by using ensemble learning [73],

n
a method for solving complicated problems by combining a z(a) = wi ai (3.11)
set of classifiers (Figure 5). In network pharmacology, especially i=1
for validation of interaction between the active molecules and
target proteins, the RF algorithm can improve the performance
Further, the result of z function is transferred to activation
of the scoring function. Chen et al. [74] combine the RF
function ∅ and output of a particular perceptron is reached:
method with network pharmacology to predict the bioactivity
of natural compounds against Alzheimer disease. RF algorithms

build different models for calculating the possibility of natural if z(a) > θ then, 1
Output = φ (z(a)) = (3.12)
compounds as a treatment option for Alzheimer’s disease. else, 0
Their findings proposed that Cynanogenin A and Methyl 3-O-
feruloylquinate has strong binding affinity with target protein
(Glycogen synthase kinase-3β). Thus, these compounds can be where θ represents a threshold parameter. Equation (3.12) esti-
used for the treatment of Alzheimer’s disease. Wu et al. [75] used mates that whether w1 a1 , w2 a2 + w3 a3 + . . . wn an − θ > 0 is a false
network pharmacology-based approach to analyze the synergistic condition or a true condition.
activity of Aconiti Lateralis Radix Praeparata. The authors used RF The architecture of MLP is composed of three layers (Figure 7):
method and built the target prediction model, which revealed that (i) input layer passes input features to the hidden layer; (ii) hidden
22 compounds have multi-target effect and can be an efficient layers may be one or more than one in MLP. The first hidden layer
candidate against incurable diseases. Similarly, Chen et al. [76] gets input features from the input layer. The first hidden layer
used RF to predict the compound–target interactions from multi- then passes the output of the perceptron to the second hidden
molecular network. Naorem et al. [77] used RF for the selection layer and so on. (iii) There is only one output layer that receives
of hub genes from pool of differentially expressed genes. After each perceptron of the previous hidden layer as an input. Where
selection, different classification models were generated using is the final node in the output layer? If the weights in a hidden
RF that confirm that the hub genes might act as biomarker layer are changed, the activation function can change the weights
and therefore easily differentiate triple-negative breast cancer in the output layer. As a result, it trains optimal weights, and
from non-triple-negative breast cancer. Similar studies in future then input features are boosted with current weights to determine
can boost the multi-target drug discovery which in turns will if a given neuron was aborted or not. Thereby, MLP employs a
provide effective treatment option against complex diseases and back-propagation approach with the activation function. As it
disorders. leverages experimental data to estimate the accuracy, therefore,
MLP has one privilege that it does not require specific structural
Deep learning information. Nowadays, researchers uses MLP to analyze the sta-
DL is a subtype of ML and is neural networks make up the bility of particular compound with target proteins [81]. Bustamam
backbone of DL algorithms, which can retrieve a higher level et al. [82] used MLP to evaluate the drug-likeness properties of
of information from input data by stacking numerous layers compounds. Similarly, Altalib et al. [83] used MLP for similarity-
together [78]. The operational workf low of DL algorithm consists based virtual screening. The future of MLP is bright, therefore,
of six main steps (Figure 6): (i) data retrivel – data are retrieved they can be proposed to expand the knowledge of compound–
from different resources; (ii) feature extraction – where it translate target interactions in network pharmacology. We believe this to
the data into useful features; (iii) data partitioning– where it is be a robust and relatively versatile model that has the potential
divided into training, testing and validation sets; (iv) developing for high generalized predictive accuracy with further tuning.
Machine learning for synergistic network pharmacology | 9

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Figure 6. Simple workf low of DL method.

DNN architecture follows the findings of a mathematical model,


which can be both non-linear or linear. Each mathematical model
was intended to be a layer, and in complicated DNN, numerous
layers are available hence, the network was dubbed as ‘deep.’
Multi-task neural networks has incorporated DNN into an integra-
tive platform so called as ‘DeepChem,’ which aids the multi-task
neural networks efficiency in drug discovery and development
[85]. Grebner et al. [86] used DNN to evaluate the bioactivity and
ADME properties of small compounds. Similarly, Chen et al. [87]
used DNN to construct gene regulatory network. Their findings
suggested that the DNN performs better and can efficiently dis-
tinguishes key functional modules between healthy individuals
and individual with coronavirus disease. These studies provide
Figure 7. Architecture of MLP. Architecture of MLP is composed of series of
layers, neurons and their connections. The inputs are forwarded through a realistic picture that DNN can be successfully used for multi-
the MLP by calculating the dot product of the input and the weights that target discovery in network pharmacology.
exist between the input layer. The hidden layer produces a value from this A CNN is used to analyze visual images [88]. CNN comprises
dot product. Transfer the estimated output from the hidden layer, then of one or more convolutional layers and is primarily used for
again calculate the dot product with the associated weights to forward it
to the next level in the MLP [Figure is adopted/modified from Dara et al.
image processing. Additionally, CNN is also considered as a regu-
[72] © (2021) Springer]. larized edition of MLP (Figure 8). CNN are completely connected
networks in which each neuron in the first layer is linked to
the neuron in the next layer. A network like CNN can over-
DNN architecture evolved from an extension of ANN and con- come the overfitting problem by employing completely integrated
tains multiple layers between input and output nodes [84]. The algorithms [89]. Hamza et al. [90] used CNN to evaluate the
10 | Noor et al.

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Figure 8. Architecture of DL. In DL, each input layers takes an input and pass to the hidden layer. The hidden layer does the activation and passes
to next hidden layer until all hidden layers are reached. Finally, an output layer is reached that gives the final outcome of the algorithm [figure is
adopted/modified from Dara et Al. [72] © (2021) Springer].

molecular bioactivities of small molecules. Their study proposed pharmacology, realistically, it is nearly impossible to collect a
that CNN yielded impressive accuracy (90.21%). Therefore, CNN significant proportion of labeled data. Similarly, choosing negative
is an efficient activity prediction approach for unknown targets. data is also challenging due to the possibility of drug interaction
In network pharmacology, the CNN model can be used to predict with more than one target. In a compound–protein interaction, if
the bioactivity of small molecules as well as to predict the binding a compound is interacting with a target protein, we considered the
affinity between bioactive compounds and target proteins, which interaction as positive. The remaining unknown interactions are
not only improves the scoring function but also lend a helping in considered to be unlabeled interactions. Most of the previously
upgrading the predictive capabilities. developed methods categorize known drug–target interactions
RNN is a special type of ANN in which the output of the as positives and a subset of unknown interactions as negatives
preceding step is used as input to the next layer [91]. Using this, a while designing a predictive model [94]. However, unknown
directed graph and a timed sequence can be generated in the net- interactions are not truly negative interactions, as they include
work. Similarly, the RNN network employs local storage to execute potential interactions that have not yet been validated as positive
groupings in input parameters. RNN was used in virtual screen- interactions. To address this problem, Xia et al. [95] developed an
ing to generate novel molecular libraries, which helped in the LapRLS that regards known interactions as positive and unknown
discovery of anticancer drugs using molecular fingerprints [92]. In interactions as unlabeled data. Chen et al. [96] developed an
network pharmacology, RNN can be used in virtual screening to algorithm using a network-based random walk with restart
generate novel molecular libraries, which helped in the discovery approach (RWRH). Unfortunately, these approaches are limited in
of multi-target drugs against diseases. When compared to other predicting the interactions of novel compounds or proteins that
traditional approaches, DL has a myriad of benefits, including the do not have any known target or drug information (e.g. newly syn-
capacity to learn complicated patterns independently. Thus, DL thesized compounds or mutated protein sequences). Additionally,
has opened new avenues for the discovery of new therapeutic collecting a large amount of data is time consuming and hard to
options for a variety of diseases and disorders. In conclusion, sig- collect.
nificant breakthroughs have been noticed in this domain, and new To deal with such data limitations, semi-supervised learning
applications and approaches are produced every day, making this (SSL) can be seen as an alternative to traditional ML. SSL uses
technique a dependable instrument in the armory accessible for extensive unlabeled data thus does not require large labeled data,
the discovery of new drug candidates. However, only few studies saving both time and cost. In SSL, source and target data come
reported the application of DL models in network pharmacology. from the same distribution. Chu et al. proposed an SSL-based
Therefore, further studies on the implementation of DL models classifier to predict the drug–drug interactions [97]. The authors
in network pharamcolgy will pave the way for multi-target drug claimed that their proposed framework showed improved perfor-
discovery in near future. mance, by up to 10.3% over the existing state-of-the-art methods
to predict drug–drug interaction prediction. Additionally, several
Challenges in training ML or DL models and their studies have used SSL to answer different biological questions [95,
potential solutions 98, 99]. Another type of naturally similar learning to SSL is active
Despite the convincing applications of ML and DL in drug–target learning where the learning algorithm can query a user interac-
interaction identification, they do have short-comings for network tively to label data with the desired outputs [100]. Active learn-
pharmacology. For example, ML model training significantly ing facilitates faster algorithm training by proactively identifying
depends on vast amounts of data [1, 93]. Model training using high-value data points in unlabeled datasets [101]. Consistent
traditional ML is also sensitive to distribution of data and with SSL, active learning does not require many labeled instances
ratio between labels and unlabeled data instances. In network and also focuses on existing unlabeled data. In active learning,
Machine learning for synergistic network pharmacology | 11

Table 1. Different learning technique that may be exploited to meet the challenges of network pharmacology

Network-pharmacology challenges Limitations of ML and DL methods Potential solutions

Small sample size ML and DL methods require a large amount of TL can be one possible solution to deal with data
data limitations. It uses previously trained model for related

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
tasks.
Large amount of unlabeled data Both ML and DL are sensitive to ratio between Semi-supervised or active learning make use of large
instances labeled and unlabeled data instances available unlabeled data instances. Hence, becoming
an effective tool for network pharmacology.
Large heterogeneous network with ML methods rely on hand-crafted features DL has the ability to simultaneously extract most
non-liner associations predictive features from complex biological networks
and learn from them. However, DL may have limited
interpretability.
Knowledge-based conditions ML and DL are data-driven Active learning involving the selective labeling could be
an option to use existing knowledge in labeling of data
instances.
Computing resources DL may require large computing resources TL can be used to mitigate the requirement of
computing resources.

the examples to be labeled are chosen carefully from large unla- TRANSFORMING NETWORK
beled data. This is contracting to passive learning where targeted PHARMACOLOGY THROUGH ARTIFICIAL
examples to be labeled are chosen randomly from unlabeled data INTELLIGENCE AND ML TECHNIQUES
[102, 103]. One major limitation of SSL and active learning is their The conclusive applications of ML and DL methods in every
dependence on a large number of negative instances, which are field of life have enhanced their popularity. To meet this surge,
hard to define in network pharmacology. several libraries and frameworks have been developed. The pur-
DL methods, as mentioned above, have also gained huge pose of developed libraries and frameworks was to ease the
popularity due to their ability to extract features from raw implementations of learning methods, thus allowing researchers
data instead of relying on hand crafted features (required in from different disciplines to apply sophisticated mathematical
traditional ML). The ability of DL methods to extract features and methods without their deeper understanding [116, 117]. Python
analyze data simultaneously is proving them as real problem- and R programming languages have been frequently used to apply
solving methods [104, 105]. However, along with large data ML or DL methods on a given task. For example, scikit-learn
requirements, DL methods have another challenge of a large [118] and Caret [119], developed in python and R, respectively,
number of parameters associated with them. Training a DL model are two commonly used libraries to apply traditional ML. Pytorch,
and determining an optimal value for it from a large number keras, tensorf low [120] are more specialized libraries for DL in
of parameters is time taking and demands huge computing python and have been widely used for training of different DL
resources that might be missing in young and emerging labs. classifiers. Pytorch is written in python programming language
In addition, interpretation from deeply trained models is difficult and enables its users to apply CNN, RNN and LSTM like familiar
[106]. deep architectures [121, 122]. Another popular framework for DL is
Transfer learning (TL) addresses the shortcomings of required tensorf low that is operable on heterogeneous operating systems.
time and necessary data amount and distribution for model Tensorf low is written in Python and C++ and provides the facility
training [107]. TL is an ML method where knowledge obtained of applying CNN, RNN, LSTM and other DL methods [123]. Keras is
from a model used in one task is reused as the foundation for another python-based library with extensive documentation and
another task [108]. TL aims for transferring knowledge across active community. It can also be utilized to apply different DL
the domains and based on similar datasets and infer relatedness architectures [124].
between these datasets using ML algorithms. TL also differs from Another integral component of analyzing big data is the scala-
SSL in terms of required distribution of data. TL is independent bility of the proposed architectures. Cloud computing has been
of distribution in input data, which is a core requirement in extensively used for many life science challenges. Cloud com-
SSL [109]. One of the most famous TL transformers for lan- puting describes both a platform and a type of application. A
guage representation models is Bidirectional Encoder Represen- cloud computing platform dynamically provisions, configures,
tations from Transformers (BERT) [110]. Language representation reconfigures and deprovisions servers as needed. On the other
models are hard to train and have been using TL frequently hand, cloud applications are extended to be accessible through
to capture [111] text semantics. BERT architecture is a multi- the Internet. These cloud applications use large data centers and
layer bidirectional transformer and has been trained on plain powerful servers that host web applications and web services
text. BioBERT [112] is biomedical version of BERT and its train- [125]. Unfortunately, the usage of distributed or parallel comput-
ing data are restricted to the biomedical domain. Both BERT ing through cloud computing is lagging behind in case of DL,
and BioBERT have been extensively used for the prediction of more specifically in biomedical sciences. Despite the availability
drug–drug interaction or drug–target interaction [113–115]. TL, of cloud-based frameworks such as Apache Spark and Elastic
when combined with deep or traditional ML, can be an effec- search, applications of cloud computing are limited in biomedical
tive tool to address the current challenges in network pharma- data sciences. Apache Spark [126] is one of the most used uni-
cology. Table 1 provides a quick overview of different learning fied analytics engines for processing large health or life science
techniques. datasets. The ease of writing code in multiple languages and
12 | Noor et al.

easily understandable structure allowed the implementation of DL method which directly operates on the graph structure. GNNs
DL architectures in the cloud. Elastic search is another framework captured the internal information directly from the graph struc-
that works for almost all types of data and has been used by ture, which means the SMILES of the molecules are transformed
leading bioinformatics research groups. However, for the majority to a graph representation of molecules. The performance of multi-
of research groups, it is quite challenging to establish cloud-based GNNs was reliable and was better than that of the ML and

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
infrastructure due to its maintenance, lack of skillful people and single GNN. The strategy of combining a global approach and a
future research thematic lines. Most of the data producing groups local approach resulted into improved model performance. The
are not fully equipped with experts in data analytics. Nvidia Clara findings of their study proposed that ZINC95618747, ZINC8577218
framework [127] provides a pre-trained model, thus eliminating and ZINC4261765 are effective inhibitors for DHODH.
the need of highly skilled experts for the discovery of drugs. It is well known that each compound has some chemi-
One major limitation of cloud-based framework is data privacy cals and physical properties including permeability, solubility,
and concerns linked to data security [128, 129]. To compensate ionization and partition coefficient which interfere with the
for such data security concerns, federated computing is rapidly pharmacodynamics and pharmacokinetics of compounds [134,
emerging [130, 131]. In federated learning, researchers or orga- 135]. Therefore, when formulating a novel drug molecule, the
nizations collaborate on their ML or DL projects without sharing physicochemical properties of compounds must be taken into
real and sensitive data, for example, patients’ data and treatment account. The physicochemical properties of chemical compounds
response in case of drug therapy. Federated computing is still has now been predicted using a variety of ML-based approaches
an emerging field and in future it will have a significant role [136]. Lusci et al. [137] introduced a framework based on a neural
in network pharmacology. Table 2 provides an overview of tools network to anticipate the molecular characteristics. Their work
available for the discovery of potential drug candidates based uses undirected cyclic graphs to classify molecules, whereas
on ML and DL algorithms. The first two columns provide a brief the earlier models for estimating physicochemical properties
description and method used behind the development of that used directed acyclic graphs. Their proposed model has ability
particular tool. The third column provides information related to to automatically extract internal representations from the
accuracy percentage of each tool, which indicates that how often molecular graphs that are well suited for the specific tasks. This
a model trained is correct. The exclusive features and limitations aspect is an important advantage for a problem like aqueous
of each tool are enlisted in the last two columns. Overall, the main solubility prediction, where the optimal feature set is unknown
advantage of providing this table is that it provides a clear picture and their number may differ from one data set to the other. It
of available ML and DL tools that can be used at different stages also saves time and avoids other costs and limitations associated
of network pharmacology pipeline. This will assist the researchers with the use of human expertise to select features. Additionally,
to integrate both ML and DL in network pharmacology for multi- various ML-based applications are widely used nowadays such
target drug discovery, which ultimately shortens time-consuming as SPARC (http://sparc.chem.uga.edu/sparc/) [138], ChemSpider
procedures and manual operations. (http://www.chemspider.com/) [139], E-DRAGON (http://www.
With recent advancements in network pharmacology research vcclab.org/lab/edragon/), OSIRIS property explorer (https://www.
techniques, it is hoped that further network-based analytical organic-chemistry.org/prog/peo/) [140], PCLIENT (http://www.
approaches would be integrated into different medical areas vcclab.org/lab/pclient/) [141], E-BABEL (http://www.vcclab.org/
to speed up the process of multi-target drug discovery. In a lab/babel/) [141] and ASNN (http://www.vcclab.org/lab/asnn/)
nutshell, the field of drug development is shifting toward network [142]. The aforementioned data support the significance of DL
pharmacology [14], but this approach faces challenges with and ML algorithms in the prediction of physiochemical properties
computational cost, time and reliability, although ML or DL offers and bioactivity of drug molecules. From a research standpoint,
promising solutions to overcome these barriers and advance the validation and preciseness of these algorithms remain a
the role of computational techniques in network pharmacology negative aspect. As a result, substantial research needs to be
(Figure 9). Thus, the combination of ML with network pharma- conducted to maximize the accuracy and reliability of AI-based
cology is considered an important topic of biomedical sciences. approaches via tailored and substantial data input. Thus, in
Regarding this, some application of ML that can be applied on network pharmacology, ML-based algorithms might help in the
network pharmacology research is discussed below in order to screening of bioactive compounds on the basis of their bioactivity
produce enthralling and promising therapeutic agents against and provide theoretic evidence for the synergistic effects of small
complex diseases. molecules for treatments of ailments.

Predicting bioactivity and physicochemical Target identification


properties of small molecules Identification of potential targets is considered as the backbone
Bioacivitiy assessment is a laborious and time-sapping process of network pharmacology research. Without foreknowledge of
in the drug discovery pipeline. Several web applications have the complete compound and disease target information, it is
recently been introduced to evaluate the bioactivity of small impossible to design effective and budget-friendly approaches for
molecules [132]. But these in silico-based analysis required bench- the treatments [143]. A huge amount of data can be obtained from
mark datasets for validation. To tackle this issue, ML approaches databases, literature and lab experiments. However, extracting
are currently used by researchers to identify the bioactivity of meaningful information from the bulk of data is a challenging
compounds which are then used to treat specific disease tar- task. To tackle this issue, ML approaches are being used for
gets. Many studies provide solid evidence of ML applications in the identification of both compound and disease-related targets.
bioactivity prediction. For example, Zhi et al. [133] used net- Laura et al. [144] introduced a multi-network-based strategy to
work pharmacology and molecular docking, which revealed dihy- integrate multi-network-based strategy to combine multiple lay-
droorotate dehydrogenase (DHODH) as a therapeutic target for ers of genomic information, particularly gene co-expression, PPI,
Small cell lung cancer. Later, they build predicted model using miRNA co-targeting and transcription factor co-targeting for iden-
Graph Neural Networks (GNN), SVM and RF. GNN are class of tification of cancer-related targets. The proposed multi-network
Table 2. The list of ML-based tools available for the discovery of potential drug candidates

Tool and software Description Method Exclusive feature Limitation Ref.

ML-based methods
QSAR-Co-X Fully accessible QSAR modeling tools for ML and classification Build a specific model equation from a variety ML requires unbiased good quality data for [195]
multiple targets. model of biological and chemical data. processing, it requires sufficient time for
Cloud 3D-QSAR An online application for developing QSAR ML Merging the sciences of molecular processing the input data with a considerable [196]
models for drug discovery. interactions, alignments and structure amount of accuracy. Other major challenges
formation are high error-openness and interpretation of
OntoQSAR An Ontology for QSAR Studies to Evaluate ML and Retrieve the biological characteristics and results. In ML, we have to run our data in all [197]
Biological and Chemical Data mathematical model chemical descriptors of active ingredients. algorithms to select the most ideal algorithm
DrugNet Using heterogeneous data to prioritize ML Drug repositioning efforts can be greatly and the selection is based on the result [198]
diseases and drugs in a network enhanced by integrating disease, drug and accuracy. This is a time taking process which
target information together. may affect the further procedure. ML
DrPOCS Drug Repositioning on the basis of Convex Machine learning With matrix completion, DrPOCS forecasts approaches are not so convenient for [199]
Sets Projection potential connections between drugs and high-level, symbolic reasoning, or planning
disorders. which creates difficulty to understand all
Chembench A hub of cheminformatics Machine learning Open platform that allows investigators to aspects of natural language [200]
access and exploit existing biological and
chemical data
mCSM-lig Structure-guided computational methodology Machine learning Giving valuable hints for assessment of [201]
for measuring the significant effect of single models, Platinum genotyping and directing drug development. It
missense mutations on the affinities of active database is efficient in detecting a variety of
compounds for proteins chemotherapeutic, antiviral and antibacterial
resistant variants
CSM-lig Online portal for predicting the binding Machine learning, Estimate binding affinities evaluate the [202]
affinity of a protein–compound complex. graph-based interaction between molecules in docked
chemical signatures complexes
based on PDBbind
databases
mCSM-AB A user-friendly web server that can estimate Machine learning Used graph-based signatures to predict [203]
antibody–antigen affinity with high accuracy antibody–antigen affinity
dendPoint Web-server for predicting and analyzing for Machine learning and Machine learning-based model capable of [204]
dendrimer pharmacokinetics principal component accurately predicting pharmacokinetic
analysis parameters
Vienna LiverTox Model used for predicting interaction between Machine learning Analyze the pharmacokinetic properties [205]
compounds and transporter of regulatory classification model
agencies
RepCOOL Repositioning of drugs computationally by Random Forest The effectiveness of the suggested technique [206]
merging diverse biological networks classifier in identifying actual drug–disease correlations
WGMFDDA A novel weighted-based Graph regularized K -nearest neighbor To uncover unidentified active [207]
matrix factorization to predict association compound–disease relationships, the graph
between compound and diseases regularised matrix factorization framework is
used. 10-fold cross-validation is carried out on
the Fdataset to assess the performance of
proposed WGMFDDA method.
Machine learning for synergistic network pharmacology
|

(Continued)
13

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
14
|

Table 2. Continued
Tool and software Description Method Exclusive feature Limitation Ref.
Noor et al.

ProTox-II Predict toxicity of bioactive compounds Molecular similarity, Predicts cytotoxicity, carcinogenicity, [208]
adsorption, distribution, metabolism and fragment hepatotoxicity, acute toxicity, mutagenicity,
excretion propensities and immunotoxicity
machine learning
ADMETlab Database for predicting adsorption, ML Systematic ADMET evaluation based on a [209]
distribution, metabolism, excretion and large and structurally diverse data sets.
toxicity (ADMET) of active compounds
Deep-learning-based
ChemGrapher Identification of chemical compounds using Deep learning Generates all the necessary information to DL lacks common sense. This makes the [210]
optical graphs link each resultant graph component to its systems fragile and when errors are made, the
corresponding source image. errors can be very large. Once trained, DL
models become inf lexible and cannot handle
multitasking. They can deliver efficient and
accurate solutions but only to one specific
problem. Even solving a similar problem
would require retraining the system.
HeteroDualNet A unique convolutional network with Convolutional Neural Used heterogeneous layers to predict the [211]
heterogeneous layers and dual branches Network compound/ drug–disease association
SAEROF To understand new features effectively Deep Neural Network Simple but effective approach to analyze the [212]
representing drug–disease associations association between compound and diseases
DeepConv-DTI Used to train, validate and test deep learning Deep learning A prediction model that efficiently improves [213]
model for prediction of compound–target the characteristics of a raw protein sequence
interaction for the purpose of recognizing local residue
patterns of target proteins produces more
improved outcome than earlier methods
DeepH-DTA Prediction of compound–target interaction Deep learning Geometrically exploits the topological [214]
structure and corresponding molecular
fingerprints of plant related compounds
DeepH-DTA Prediction of compound–target interaction Deep learning Geometrically exploits the topological [214]
structure and corresponding molecular
fingerprints of plant related compounds
IDDkin Identification of kinase inhibitors Deep diffusion model In silico method integrating data from [215]
different resources to predict the kinase
inhibitors
Quantum-Based
COSMOfrag a brand-new method based on quantum chemistry for Used quantum chemistry for predicting the [216]
high-throughput adsorption, distribution, metabolism and excretion adsorption, distribution, metabolism and
property prediction Quantum Chemistry excretion properties of plant related
compounds

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Machine learning for synergistic network pharmacology | 15

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Figure 9. Advancing network pharmacology research via ML. A superpower to fight against the deadliest diseases. DL CNN, DNN, RNN and MLP holds
promise for drug development as it allows the prediction of molecular structure and function, and the automated development of novel chemical entities
with specific features. Following that, SVM and RF has spotlighted recently as they significantly applied to handle big data challenges in network
pharmacology research. Thus, ML approaches facilitate the development of drugs with multi-target effects and exploring the poly-pharmacology of
different currently used drugs.

strategy is perfectly suited to identify these potential miRNA identification of compound and disease-related targets using ML
drivers since one of the layers is exactly the miRNA co-targeting technqiues can be helpful for researchers to get a better under-
network and, in this case, the role of the other layers is to filter standing of targets.
among all the miRNAs only those whose targets also interact in
one or more of the other layers and in particular in the cancer
co-expression one. The authors used consensus clustering algo- Identification of molecular pathways
rithm of ML on the predicted network which uncovered CD46, Pathways analysis is a crucial step in network pharmacology as it
BTG2, ATF3, HDGF and F11R as driver cancer genes. In short, is intended to give some special meaning to high-throughput bio-
such kind of multi-network-based analysis provide encouraging logical data [147]. Despite their extensive usage, the context and
evidence that multilayer networks contain new information on fundamentals of pathway analysis may not be fully understood,
the structure and dynamics of complex systems, which would not which ultimately led to false results [148]. To overcome these
be possible to get from the individual components. Beyond this, problems, ML are extensively used for pathway analysis. Inte-
group of researchers proposed DEcRyPT method for multi-target gration of ML techniques with pathway analysis provides a new
drug discovery [145, 146]. DecRyPT method used the RF algorithm insight for predicting the molecular targets of multiple diseases
that contributed to a preferential exploration of disease-related and disorders. STRING [149], STITCH [150] and DisGeNET [151]
targets. Consequently, ML models offered a quantitative frame- are widely used in network pharmacology for the identification of
work to identify the promising targets for discovery of novel drug molecular pathways in which dysregulation leads to disease con-
candidates. In the context of network pharmacology, researchers ditions. In short, the birth of ML in pharmaceutical research has
rely on literature and databases for target identification. However, made it possible to find molecular targets or pathways of human
16 | Noor et al.

diseases using genomic data. A number of techniques for discov- evolved in 2011 to prefigure the PPIs based on frequency counts,
ering (or designing) novel pathways have been proposed, including domain-domain interactions and ML. Beyond this, Chen et al.
search-based methods which identify plausible paths between [160] developed an ensemble learning approach for PPI predic-
given input and output metabolites. Other approaches include tion that integrated multiple learning algorithms and different
searching for frequently-occurring patterns of molecular func- protein-pair representations. The authors adopted a stacked gen-

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
tions in biological networks or kernel-based methods for learning eralization scheme and develop a classifier called PPI-MetaGO,
associations between enzymes catalyzing successive functions which improves PPI prediction by deducing the biases of the
in metabolic pathways [152]. Dongardive et al. [153] used ML base generalizers and exploiting the synergy among various ML
classifier techniques for pathway identification. The authors used algorithms. Many examples of ML role in PPI prediction has been
five different ML classifier to detect molecular pathways involved discussed, and the PPIs research is still in progress. However,
in viral infections and cancer. The authors applied five different designing PPI modulators continues to be a tricky problem, and
ML classifiers to determine the accuracy of the classification. The is typically endorsed by a comprehensive understanding of the
overall results showed that ML classifiers could be able to give a system at the molecular and structural levels. In terms of network
range of accuracy of 71–72%. Overall, RF gave the highest accuracy, pharmacology, PPIs networks are being used more frequently
which indicates that RF technique and the proposed method as universal platforms for the analysis and integration of data.
might be efficient in classifying the cancer and viral metabolic Therefore, these models can be implemented in network phar-
pathways. Thus, RF methods can be used in network pharmacol- macology, which provides a helping hand in multi-target discov-
ogy for identification of metabolic pathways. Similarly, Hao et al. ery. Altogether, gaining a better understanding of PPIs using ML
[154] introduced PASNet, the first DNN that presents a hierarchi- approaches will allow researchers to unveil the therapeutically
cal representations of genes, pathways and their nonlinear effects. relevant aspects of PPIs to treat human diseases.
PASNet models a multi-layered, hierarchical biological system of
genes and pathways to predict clinical outcomes by leveraging DL. Network analysis
The sparse solution of PASNet provides the capability of model Recent studies on biological networks, including metabolic, gene
interpretability that most conventional fully-connected neural regulatory and PPI networks, have contributed to understanding
networks lack. In the context of network pharmacology, the use the origins of cellular behaviors and evolutionary design princi-
of PASNet can provide a better understanding of metabolic path- ples, as well as specific cell biological processes or diseases [161].
ways. Although ML-based approaches play key roles in construct- In terms of network pharmacology, network analysis provides
ing and understanding metabolic pathways and their subparts, novel insights in understanding the interactions among biologi-
the prediction and construction of synthetic metabolic pathways cal entities, such as bioactive compounds, targets, diseases and
is a significant challenge in network phramacology research. pathways [162]. In pharmacological studies, multiple active com-
pounds can target one gene, and one compound can target mul-
Prediction of PPIs tiple genes, which necessitates the construction of compound–
PPIs are the center of network pharmacology. PPI analysis is essen- target and compound–target–pathway networks [163]. However,
tial for designing efficient drugs as it plays a key role in the cellular constructing such networks is challenging and prone to high
systems of all living organisms [155]. Large PPI data produced by false-positive rates. Therefore, extracting key sub-networks and
high-throughput techniques gave boost to a new drug discovery identifying overlapping networks is crucial to identify the key
era that ultimately became the basis of new biological discoveries. components in the network.
Managing, evaluating and modeling PPI data is a primary chal- ML-based algorithms are effective method to process the bio-
lenge in network pharmacology [156]. These issues are currently logical network data, so as to implement classification, cluster-
addressed by ML approaches which uses various data sets, input ing and prediction tasks in a biological network. Therefore, ML
attributes and architectures to predict PPIs. Furthermore, recent can effectively predict the multitarget mechanisms underlying
studies claim that DL is the most effective approach for predicting the effects of active compound against diseases. Recently, Zeng
PPIs [157]. et al. [164] introduced an deepDTnet for target identification from
Hashemifar et al. [136] introduced DPPI (https://github.com/ heterogenous networks. DeepDTnet computationally identifies
hashemifar/DPPI/), a novel DL framework for predicting PPI. DPPI thousands of novel drug–target interactions with high accuracy
used only sequence information and then predict interactions by using DNNs. DeepDTnet learn low-dimensional vectors rep-
between proteins. DPPI uses CNN model and effectively mea- resentations for both drugs and targets by a unique integra-
sures interactions by combining pre-existing experimental PPI tion of large-scale chemical, genomic and phenotypic profiles.
data and evolutionary information of protein pair under predic- Owing to the lack of experimentally reported negative samples
tion. Du et al. [158] proposed a cutting-edge method known as (non-interactions between drugs and targets) from the publicly
DeepPPI (http://ailab.ahu.edu.cn:8087/DeepPPI/index.html) that available databases, deepDTnet employed the Positive-Unlabeled
uses DNN to effectively learn the representations of proteins from (PU)-matrix completion algorithm to low-rank matrix completion,
common protein descriptors. The experimental results demon- which is able to infer whether two drugs share a target without
strate that DeepPPI outperforms on the test data set, specificity negative samples as input. Similary, Zheng et al. [165] developed
of 94.49%, recall of 90.56%, precision of 94.38% and accuracy THN_KRLS, an ML-based prediction method for tripartite hetero-
of 92.50%, respectively. One factor that contributes to the good geneous network. THN_KRLS combining multiple kernels into a
performance of DeepPPI that it can learn useful features of pro- tripartite heterogeneous drug–target–disease interaction spaces
teins pairs by a layer-wise abstraction. Another factor is the in order to integrate multiple sources of biological information
DeepPPI can learn an internal distributed feature representation simultaneously. This novel network algorithm extends the tradi-
automatically from the data. These characteristics are proved tional drug–target interaction bipartite graph to the third disease
to be an effective approach to learning representations of pro- layer. Following that, ML can be implemented in network phar-
teins. Further, PPI_SVM [159] (http://code.google.com/p/cmater- macology not only for efficient handling of high throughput, het-
bioinfo/), an efficient knowledge-based prediction method, was erogeneous and complex molecular data, but also for mining of
Machine learning for synergistic network pharmacology | 17

feature or relationship in the ‘compound-target’ and ‘compound- top classes of drugs, along with their scatter plot. This can help
target-pathway’ network. In the context of network pharmacol- scientists to predict the action mechanisms and can help them
ogy, these kind of ML algorithms will help in predicting affinity in the discovery of new drugs. Gao et al. [180] developed Genetic
values within ‘compound-target’ network which will explore more profile–activity relationship (GPAR) to help modeling and predict-
core compounds and core target. To sum up, the ML model lend a ing action mechanisms easily via DL. The users can use GPAR

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
helping hand in the identification of multi-target compounds for to customize their training sets to train self-defined predictions
given target proteins based on their involvement in pathways in models, to evaluate the model performances and to make further
which dysregulation leads to disease conditions. predictions automatically. Cross-validation tests show GPAR out-
performs gene set enrichment analysis in predicting mechanism
Hub gene analysis of actions. Similarly, Liu et al. [181] and Fronsdal et al. [182] used
Topological analysis of PPI network is used to identify hub genes DL models and predict action mechanisms of compounds. Their
related to disease. Hub genes are those genes having high degree studies highlighted the importance of DL models for understand-
of connectivity in gene network, suggesting functional impor- ing the action mechanisms of novel drugs. Implementation of
tance in the diseases [166]. High connectivity means that the DL models in network pharmacology for predicting toxicity and
connectivity ranked at top 10%. As for example, if the size of actions mechanism might reveal efficacy and synergistic effect of
module is 10,000, then the top 1000 genes are considered as compounds.
the hub genes [167]. Therefore, accurate identification of hub
genes is crucial to design multi-target drugs. Nowadays, ML with Integrating ML systems for improving docking
its powerful models are used to predict hub genes from gene prediction reliability
networks. Recently, Liu et al. [168] identified nine-hub-gene sig- In network pharmacology, the pressing need of the hour is to
nature of metabolic syndrome using integrated bioinformatics grasp poly-pharmacological effects of compounds and to develop
analysis and RF method. This nine-hub-gene signature showed computational algorithms to accurately predict the effect of com-
better calibration and classification performance (AUC = 0.861 in pounds across the molecular network. To achieve this, high-
external validation set, AUC = 0.968 in training set and AUC = 0.883 throughput molecular docking simulation systems must be con-
in internal validation set). Similarly, Li et al. [169] applied RF meth- structed to calculate the combined effects of compounds through
ods to identify the stromal-related features of the two modules molecular networks. ML is often presented as a new industrial
at a single-gene level. Their study proposed 71 hub genes that revolution. The sampling, scoring and computing time are three
can be used as a biomarker for esophageal adenocarcinoma and main challenges of molecular docking. Nowadays, researchers
squamous cell carcinoma. Spotting the benefits provided by ML are using ML algorithms to overcome these challenges. Recently,
approaches for hub gene analysis, we strongly believe that these Google’s sister company, DeepMind, in London, developed an ML-
ML approaches can be used in network pharmacology for the based tool named as AlphaFold for predicting the structure of
identification of hub genes which not only provide promising target protein [183]. Following that, another tool RoseTTAFold has
therapeutic target of disease but also accelerate the process of been developed that uses DL to quickly and accurately predict
multi-target drug discovery. the protein structures based on limited information [184]. After
successfully predicting the 3D structure of target protein, docking
Prediction of the mode of action and toxicity of analysis is performed to predict the binding affinity between
compounds compound and target proteins. In ML, a model is trained using
Compound toxicity is the degree to which active ingredients or training data (active/inactive compounds). The trained model is
a particular chemical substance can damage an organism. With then tested for accuracy, and if it passes muster, it is applied to
the advent of ML techniques, the resources, time and cost of fresh datasets to figure out compounds for desired activity against
designing candidate drugs is decreased to an unprecedented level. a target [185]. The finalist compounds are then undergo ADMET
ML offers a helping hand by developing multiple tool/software to analysis, including several bioassays prior to entering in clinical
determine the toxicity of compounds including Tox21 (https:// research.
ntp.niehs.nih.gov/whatwestudy/tox21/index.html) [170], Toxtree In network pharmacology, target docking is used for validation
(http://toxtree.sourceforge.net/) [171], admetSAR (http://lmmd. of interaction that exists between target and compound. Regard-
ecust.edu.cn/admetsar2/) [172], TargeTox (https://github.com/ ing this, various ML algorithm including SVR-Score, NNScore, ID-
artem-lysenko/TargeTox) [173], SEA (http://sea.bkslab.org/) [174], score and CScore has now been developed for target docking
eToxPred (https://www.brylinski.org/etoxpred-0) [175], pkCSM [186]. In the same vein, many other algorithms CNNs, RFs, shallow
(http://biosig.unimelb.edu.au/pkcsm/) [176] and LimTox (http:// neural networks and SVM are used for the prediction of binding
limtox.bioinfo.cnio.es/) [177]. Mayr et al. [178] developed DeepTox affinity between protein-compound. Kwon et al. [187] introduced
pipeline for toxicity prediction. DeepTox first normalizes the AK-score that predicts protein-ligand binding affinity based on
representation of chemical compounds. Next, it computed the ensemble model of three-dimensional neural network. Sim-
chemical descriptors that are used as input in ML techniques. ilarly, Amangeldiuly et al. used SVM and RF to predict binding
After that, it build models and then trained the model. Later, the affinity between protein and ligands. Qamar et al. [188] reported
performance of the trained model are then evaluated. Finally, the that target-specific ML scoring function has better performance
well-trained model is used to predict the toxicity of the active compared to the classical generic scoring function in ranking
compound. biologicaly active molecules against the target enzymes during
Understanding a biologically active compound’s mechanism of drug discovery. Hsin et al. [189] combined ML approaches with
action entails not only identifying the target but also investigating docking simulation to improve the reliability of docking analysis
the biological chemistry that occurs before or after target binding. in network pharmacology. In summary, the implementation of ML
Gururaj et al. [179] introduced a web application through which a techniques makes its simpler with high accuracy and precision.
user can input a custom testing features data-set containing gene The combination of ML techniques with network pharmacology
expression and cell viability levels. The output produced is the shows a remarkable advantage in highlighting the systematic
18 | Noor et al.

biological network. With ML, network pharmacology has trans- novel applications of DL and ML algorithms continuously being
formed into a potent tool for interpreting the biologically active developed.
compounds and possible targets of herbal remedies.

Clinical trials ONGOING CHALLENGES IN ADOPTING ML:


LEADS ON WAYS TO OVERCOME THE

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
ML have the potential to add much value to a clinical trial by
facilitating decision-making, reducing the time to complete the LIMITATION OF NETWORK
trial, and the overall drug development process. ML approaches PHARMACOLOGY
are available to analyze large and heterogeneous data, identifying Drug research benefits from network pharmacology will aid in
intricate and occult patterns and predicting complex outcomes. the resurgence of traditional knowledge. Despite that, there are
As a result, ML has value to add across the spectrum of clin- several drawbacks of adopting ML in network pharmacology that
ical trials, from preclinical drug discovery to pre-trial planning would hopefully get resolved in the future. First, the availability
through study execution to data management and analysis. For of a large amount of data is critical to the success of ML because
example, a treatment that is effective against early manifestation these data are used for the subsequent training provided to the
might not be effective against late manifestation. In such cases, system. Secondly, a corporation may incur additional fees when
Heterogeneous Treatment Effect (HTE) analysis can be extremely accessing data from several database. Thirdly, data must also be
useful in finding subgroups consisting of patients who have sim- trustworthy and of good quality to provide accurate result predic-
ilar covariates and display similar treatment responses. In the tion. Many other barriers to full-f ledged ML adoption in network
context of a clinical trial, HTE analysis can increase the likelihood pharmacology included availability of qualified professionalist
of identifying subgroups of the population for whom a particular to operate AI-based platforms, insufficient resources for smaller
treatment is effective, even when it is found to be ineffective companies, fear of replacing workers, which could result in a loss
for the population as a whole. Using ML, Rouillard et al. [190] of jobs, uncertainty about the data generated by ML and the black
assessed omics data for a set of 332 targets that succeeded or box phenomenon.
failed phase III clinical trials by multivariate feature selection. With the passage of time, specific tasks related to drug devel-
They found gene expression data were particularly predictive of opment, production, clinical trials, supply chains and sales will
successful targets, characterized by low mean RNA expression be automated; however, these all tasks fall under the bracket
and high variance across tissues. This study confirmed previous of ‘narrow ML,’ in which ML must be trained by bulk of data
findings that ideal targets exhibit disease-specific expression in in order to be appropriate for a specific activity. Thus, human
affected tissues. Bisaso et al. [191] observed a steady growth involvement is crucial for effective designing and impelmenta-
in the application of ML in HIV clinical research. Despite the tion of the ML platform in network pharmacology, although ML
dominance of statistical analysis as an application paradigm, ML is already replacing monotonous jobs, which leaves plenty of
usage is steadily diversifying to include practical clinical tools room for incorporating human intelligence for exploring more
to assist in clinical decision making. The use of high-dimension sophisticated insights and creativity in the field of drug devel-
sources of data such as genomic data has overtaken other data opment. To date, bioinformatics is gaining popularity in every
sources and this has been attended by an increase in use of aspect of the life sciences [192]. One of the major thrusts of
high performance non-parametric ML methods with a focus on current bioinformatics approaches is the prediction and identifi-
combating resistance to antiretroviral therapy. To the extent that cation of biologically active candidates and mining and storage of
ML-enabled clinical research can improve the efficiency and qual- related information [193]. Another interesting aspect of bioinfor-
ity of biomedical evidence, ML innovation has given new essen- matics is the ability to utilize high-throughput data and computa-
tiality to network pharmacology. With more clinical data and tional methods to predict potential synergistic drug combinations
with availablity of more reboust ML methods, it is expected that [194]. Following that, bioinformatics in coordination with ML can
ML-based approaches will have significant impact onnetwork help to streamline the drug discovery process and increase the
pharmacology and will become the standard computer-supported chances of success by identifying combinations of drugs that
medication plan strategy. have the potential to produce a more potent therapeutic effect
To sum up, the ML models may be different each time they are while reducing toxicity. Further, current bioinformatics methods
trained. In turn, the models may make different predictions, and help in addressing the challenges posed by large amounts of
when evaluated, may have a different level of error or accuracy. unlabeled data in network pharmacology by leveraging compu-
The amount of difference in the results will be related to how tational methods to organize, process and analyze vast amounts
different the training data is for each model, and the variance of data. Overall, with the development of innovative computa-
of the specific model and model configuration chosen. A more tional analysis combined with the increasing amount of relevant
sensitive algorithm has a larger variance, which will result in biomedical information, it could be the ‘perfect storm’ for bioin-
more difference in the model and, in turn, the predictions made formatics to address some of the challenges faced by network
and evaluation of the model. Conversely, a less sensitive algo- pharmacology to discover efficacious multi-target drugs of the
rithm has a smaller variance and will result in less difference future.
in the resulting model with different training data and, in turn, Untill now, only a few network pharmacology-based studies
less difference in the resulting predictions and model evalua- used ML at some specific stages particularly for compound-
tion. Thus, the ML model is making different predictions each related target prediction and diseases-related target prediction.
time it is trained, even when it is trained on the same data set However, network pharmacology is lagging behind in the race
each time. Table 3 enlists the ML methods used at each step of of using ML for developing more entrailing treatment options
network pharmacology, their general workf low with their short against deadliest diseases and disorders. Despite mentioned
description. The methods mentioned here capture many of the limitations, the combination of ML approaches with network
key areas of how ML and DL can inf luence the multi-target drug pharmacology is warmly welcomed by the scientific community
discovery. Nonetheless, this is still an active area of research with because it provides a new insight to the esoteric knowledge
Table 3. Application of ML and deep learning principles in network pharmacology for multi-target drug discovery

Sr. No# Steps Methods/tools General workflow Description

1 Predicting Lusci et al. [137] • This framework employed Undirected Graph Recursive Neural Networks • The UGRNN method shown to be able to build predictive solubility
bioactivity and framework (UGRNN) which first transforms molecular structures into vectors of the same models that were comparable in accuracy to models built with molecular
physicochemical length as the molecular representation and then passes them to a fully descriptors.
properties of connected neural networks layer to build models for prediction
small molecules

Zhi et al. [133] • Graph neural networks (GNNs) used to build reliable predicted models for • The proposed pipeline identifies novel targets related to dihydroorotate
study discovering new dihydroorotate dehydrogenase protein inhibitors for small cell dehydrogenase (DHODH) and to screen drug candidates for their potential
lung cancer to inhibit multiple targets in small cell lung cancer.

2 Target Cantini et al. • This network-based workflow used an expression dataset supplied by the user, • The combination of the different layers of information allowed us to
identification [144] creates the four-layer multi-network [(i) transcription factor (TF) co-targeting extract from the multi-network indications on the regulatory pattern and
network-based network, (ii) microRNA co-targeting network, (iii) protein–protein interaction functional role of both the already known and the new candidate driver
biology analysis (PPI) network and (iv) gene co-expression network)] and provides as output the genes.
workflow multi-network community structure
• The authors applied a consensus clustering algorithm (An ML-based biology
analysis algorithm that divide the network into sub-modules with different
functions) on identified network communities to discover cancer driver genes

Drug–Target • DEcRyPT method uses regression RF as an orthogonal learning approach to • DEcRyPT can effectively advance early natural product-based drug
Relationship self-organizing maps. It harnesses a user-defined number of predictors discovery, and can be a swift alternative to chemoproteomics and work
Predictor (decision trees) that independently analyze different portions of the training as a general strategy for systems pharmacology studies.
(DEcRyPT) [146] data, before aggregation of the resulting predicted outputs.
method • When applied to celastrol, DEcRyPT method unveiled modulation of
cannabinoid receptors by celastrol, which are implicated in cancer progression

3 Identification of Pathway- • PASNet builds a network model by leveraging prior biological knowledge of • Pathway-Associated Sparse Deep Neural Network (PASNet) outperformed
molecular associated pathway databases and by taking hierarchical nonlinear relationships of other models to predict the outcome of glioblastoma multiforme (GBM)
pathways sparse Deep biological processes into account with an AUC of 0.66
Neural Network • To improve the model interpretability, PASNet introduced sparse coding to
(PASNet) [154] capture significant components of a biological mechanism in the model

Dongardive • The author collected data from metabolic networks of six different types of • Pathway-based RF classification allows us to learn more from the
et al. [153] cancer pathways and used different ML classifier to determine the accuracy of biological system rather than looking at individual genes, examine how
method the classification for selected datasets various pathways are interconnected and investigate the shared
components among different pathways

4 Prediction of DPPI [136] • DPPI divided a protein into subsequences, and used these subsequences to • DPPI efficiently model and predict PPIs from sequence and is generalizable
protein–protein perform multiple sequence alignment through PSI-BLAST to obtain a protein to many applications including the prediction of model binding affinities
interactions (PPI) profile. This is then fed to a convolutional module and a random projection
module to predict PPIs

DeepPPI [158] • DeepPPI used a Deep Neural Network architecture network for each protein to • DeepPPI capture informative features of protein pairs by a layer-wise
extract high-level discriminative features from common protein descriptors abstraction.
• This method comprises two different architectures: DeepPPI-Sep, which uses • In addition, DeepPPI can automatically learn an internal distributed
two separate networks as input for each protein, and DeepPPI-Con, which feature representation from the data.
directly links two proteins in a single network

PPI_SVM [159] • PPI_SVM uses SVM to predict interactions between two protein sequences by • PPI_SVM not only consider all possible interactions between constituent
exploiting their domain information. domains of interacting protein pairs but also determining interaction
Machine learning for synergistic network pharmacology

affinity between a protein pair by distinguishing the dominant interacting


|

domain pair
(Continued)
19

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Table 3. Continued
20
|

Sr. No# Steps Methods/tools General workflow Description

PPI-MetaGO [160] • PPI-MetaGO constructs a meta-classification model for PPI prediction using • PPI-MetaGO improves PPI prediction by deducing the biases of the base
three types of features: physicochemical features, gene ontology-term features generalizers and exploiting the synergy among various ML algorithms.
and network-based features.
Noor et al.

5 Network DeepDTnet [164] • DeepDTnet first build a heterogeneous network connecting drugs, targets and • Additionally, deepDTnet shows higher robustness in comparison RF, SVMs,
analysis diseases by integrating 15 types of chemical, genomic, phenotypic and cellular k-nearest neighbors and naive Bayes, since it performs well on drugs or
network profiles targets showing high and low connectivity as well as high or low chemical
• DeepDTnet then learn low-dimensional but informative vector representations similarity.
within network for both drugs and targets using a Deep Neural Networks model
for graph representations (DNGR) algorithm
• For any given drug–target pair, it is difficult to verify unobserved evidence that
such a connection is, indeed, nonexistent, or hidden, therefore, deepDTnet
employ the positive unlabeled-learning formulation to low-rank matrix
completion, which is able to infer whether two drugs share a target or not
THN_KRLS [165] • THN_KRLS adds a disease layer to the traditional drug–target interaction • THN_KRLS method performs better than the other existing methods
bipartite graph. when known drug–target interactions are missing in the training data
• From the constructed heterogeneous network, new drug–target interactions can
be inferred with Gaussian kernel functions and the regularized least square
method of the Kronecker product.

6 Hub gene RF algorithm • Li et al. applied RF algorithm to estimate the contribution of differentially • RF has ability efficiently predicts hub genes from pool of differentially
analysis [168, 169] expressed genes in esophageal carcinoma. expressed genes.
• Similarly, Liu et al. used LASSO regression and RF methods for hub gene feature
selection. The author selected final genes based on the classification accuracy of
ML approaches.

7 Prediction of the DeepTox [178] • DeepTox normalizes the chemical structures followed by computation of the • DeepTox supported the automatic learning of features that were similar
mode of action chemical descriptors. to well-established toxicophores identified by expert knowledge and
and toxicity of • The computed descriptors are used in DL methods to predict the toxicity of experience.
compounds chemicals.

Flask-based web • Neural network model is integrated into the web application through which a • Flask-based web application can help researchers to predict the
application [179] user can input a custom testing features data-set containing gene expression mechanism of action and can help them in the discovery of new drugs.
and cell viability levels.
• The output produced is the top classes of drugs, along with their scatter plot.
Genetic • GPAR assists in modeling and predicting mechanism of action through DL. • GPAR offers a more powerful connection of expression signatures and
profile-activity • The users can use GPAR to customize their training sets to train self-defined action mechanism, which could get the high accuracy in modeling action
relationship mechanism of action prediction models, to evaluate the model performances mechanisms and querying signatures, and facilitate drug repurposing
(GPAR) [180] and to make further predictions automatically. opportunities.
8 Integrating ML Kwon et al. [187] • AK-Score utilizes independently trained networks to make predictions and the • AK-Score is a new neural network model for binding affinity prediction
systems for average of those values is considered as the final prediction. of a protein–ligand complex structure
improving Qamar et al. [188] • The author used ML approach to set up a pipeline that scores and ranks • Their findings demonstrates that ML improve the accuracy of
docking biological molecules against the 3CLpro enzymes. structure-based virtual screening by promoting the identification of most
prediction • The RF classifier was used to predict molecule class and RF regression to potential ligands having high accuracy score
reliability determine the molecule’s activity

Hsin et al. [189] • The author combined multiple docking tools and two ML systems to predict the • Integrating this approach with molecular network maps makes it
high-precision binding affinity of redocked ligand poses. possible to address drug safety issues by comprehensively investigating
computational • The first ML system include, not only intermolecular interactions, but also network-dependent effects of a drug or drug candidate.
prediction exploited the physicochemical properties of the ligand as additional features.
approach • The second ML system helps in the selection of most predictive binding modes
for each of the complexes.

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Machine learning for synergistic network pharmacology | 21

of traditional medicines. Therefore, there is an urgent need to pharmacology but additional studies are crucial in understudied
combine ML approaches with network pharmacology to achieve areas, generating ML-based pipeline for multi-target drug discov-
better and more accurate outcomes. ery (Supplementary Figure S1). ML outputs are highly dependent
This literature review has some limitations that must be kept on the initial values or weights of the network parameters or even
in mind for further research. There are some important research the order in which training examples are presented to the net-

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
questions that might give important information respect to the work. The availability of high-quality, accurate and curated data in
trends and challenges of incorporating ML in network pharma- large quantities to train and develop ML models is also challeng-
cology. For example, we do not specify that which features should ing. The requirements for the amounts and accuracy desired are
include in ML models to foster the multi-target drug discov- dependent on the complexity of the data type and the question
ery through network pharmacology. Secondly, this study does to be resolved. Thus, it can be expensive to generate these data
not discuss the automated pipeline that integrate network phar- sets. Pre-competitive consortia of pharmaceutical companies and
macology with ML for multi-target drug discovery, as only a academic institutions that use appropriate data standards and
few network pharmacology-based studies used ML at some spe- have the necessary operational and open data frameworks may
cific stages particularly for compound-related target prediction be part of the solution to meet these data demands. Though there
and diseases-related target prediction. Therefore, further research are some unavoidable obstacles and tremendous amount of work
should include other analysis including compound–target protein has to be done to incorporate ML tools in network pharmacology,
interaction network analysis as well as compound–target protein– there is no doubt that in the near future ML will bring revolu-
disease interaction network analysis in order to provide a more tionary changes in network pharmacology for multi-target drug
complete understanding of the impact of ML on multi-target drug discovery.
discovery. To sum up, further research needs to be done to give
an answer to the aforementioned issues in order to provide other
directions and continue to enlarge the knowledge about ML in
network pharmacology. Besides, with the intention of ensuring the Key Points
accuracy of the ML models, it is imperative that further research • Network pharmacology has become an important
includes big data. research model for multi-target drug discovery. However,
there is a need to improve the predictive performances
in network pharmacology.
CONCLUDING REMARKS AND FUTURE • The recent successes of the machine/deep learning
PERSPECTIVES (ML/DL) approaches in drug discovery could be used to
The pharmaceutical industry experienced an unprecedented rise overcome the limitations of network pharmacology.
in the cost of developing new drugs. Society needed specific • With the increasing number of ML available libraries,
major improvements in this area as the healthcare industry cur- toolkits and frameworks, we expect to see additional
rently faces a number of complicated challenges, such as the research studies and also the examples of ML- or DL-
skyrocketing costs of both therapies and drugs. Regrading this, based production pipelines for network pharmacology in
network pharmacology is considered as a new frontier in drug the near future.
discovery. Network pharmacology is an in silico approach, inte- • This review provides an overview of ML and DL
grating bioinformatics and chemoinformatics that helps to build approaches that can be opted to develop multi-target
a network-centric view of drug action. The network pharmacol- drugs.
ogy technique lays the most recent scientific groundwork for
evaluating the effectiveness of multi-component, multi-target
compound formulae and investigating the disease treatment of
multiple therapeutic targets. The ongoing implementation of ML SUPPLEMENTARY DATA
and its tools promises to improve discovery and decision making
Supplementary data are available online at https://academic.oup.
in network pharmacology. In the light of the current study, we
com/bib.
thoroughly discuss the backend of ML approaches in supporting
network pharmacology research along with its limitations and
opportunities for the pharmaceutical industry. This assembled ACKNOWLEDGEMENTS
data aimed to discuss the role of ML-based approaches in network
Authors would like to acknowledge Integrative Omics and
pharmacology for providing more promising treatment options.
Molecular Modeling Laboratory, and Biomedical Data Science
We have attempted to cover this vast topic in this review and
Laboratory, Department of Bioinformatics and Biotechnol-
hope that it will serve as a reference for clinicians who are eager
ogy, Government College University, Faisalabad, Pakistan, for
to bring advances in network pharmacology for multi-target drug
providing necessary support and facilities to conduct this
discovery.
study.
For the future, we believe that the combination of advanced ML
systems with network pharmacology could make an encouraging
progression in multi-target drug discovery. The field is open to
FUNDING
explorations of how active compounds and target proteins can
be used by ML algorithms for concrete drug discovery appli- This research received no specific grant from any funding agency
cations. Despite the fact that both network pharmacology and in the public, commercial, or not-for-profit sectors.
ML has provided many novel targets and unique compounds,
there has not yet been any success story in which compounds
and target proteins identified through network pharmacology
DATA AVAILABILITY
and ML made it to the market for general usage. There is no The data underlying this article are available in the article and in
doubt that ML will soon bring dramatic improvements in network its online supplementary material.
22 | Noor et al.

REFERENCES epimedium for alleviating mild cognitive impairment and


treating Alzheimer’s disease. Evid Based Complement Alternat
1. Vamathevan J, Clark D, Czodrowski P, et al. Applications of
Med 2021;2021:2302680.
machine learning in drug discovery and development. Nat Rev
22. Niemira M, Collin F, Szalkowska A, et al. Molecular signature
Drug Discov 2019;18:463–77.
of subtypes of non-small-cell lung cancer by large-scale tran-
2. Hughes JP, Rees S, Kalindjian SB, Philpott KL. Principles of early

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
scriptional profiling: identification of key modules and genes
drug discovery. Br J Pharmacol 2011;162:1239–49.
by weighted gene co-expression network analysis (WGCNA).
3. Sinha S, Vohora D. Drug discovery and development: an
Cancer 2020;12:37. https://doi.org/10.3390/cancers12010037.
overview. In: D. Vohora, G. Singh (eds.). Pharmaceutical Medicine
23. Liang X, Li H, Li S. A novel network pharmacology approach to
and Translational Clinical Research. Boston, FL: Academic Press,
analyse traditional herbal formulae: the Liu-Wei-Di-Huang pill
2018, 19–32.
as a case study. Mol Biosyst 2014;10:1014–22.
4. Jørgensen JT. A challenging drug development process in the
24. Ren Y. Research progress and challenges of network pharma-
era of personalized medicine. Drug Discov Today 2011;16:891–7.
cology in field of traditional Chinese medicine. Chin Tradit Herb
5. Lu JJ, Pan W, Hu YJ, Wang YT. Multi-target drugs: the trend of
Drug 2020;18:4789–97.
drug research and development. PloS One 2012;7:e40262.
25. Li S. Network pharmacology evaluation method guidance-
6. Mátyus P. Multi-targeting drugs: past, present and future. Orv
draft. World J Tradit Chi Med 2021;7:148.
Hetil 2020;161:523–31.
26. Jordan MI, Mitchell TM. Machine learning: trends, perspectives,
7. Schrattenholz A, Groebe K, Soskic V. Systems biology
and prospects. Science 2015;349:255–60.
approaches and tools for analysis of interactomes and
27. Patel L, Shukla T, Huang X, et al. Machine learning methods in
multi-target drugs. Methods Mol Biol (Clifton, NJ) 2010;662:29–58.
drug discovery. Molecules 2020;25:5277. https://doi.org/10.3390/
8. Bebek G, Koyutürk M, Price ND, et al. Network biology methods
molecules25225277.
integrating biological data for translational science. Brief Bioin-
28. Hoffmann CH. Is AI intelligent? An assessment of artificial
form 2012;13:446–59.
intelligence, 70 years after Turing. Technol Soc 2022;68:101893.
9. Diaz-Beltran L, Cano C, Wall DP, et al. Systems biology as a
29. Robbins S. A misdirected principle with a catch: explicability
comparative approach to understand complex gene expression
for AI. Mind Mach 2019;29:495–514.
in neurological diseases. Behav Sci 2013;3:253–72.
30. Jiménez-Luna J, Grisoni F, Weskamp N, et al. Artificial intelli-
10. Chen BS, Wu CC. Systems biology as an integrated platform
gence in drug discovery: recent advances and future perspec-
for bioinformatics, systems synthetic biology, and systems
tives. Expert Opin Drug Discovery 2021;16:949–59.
metabolic engineering. Cell 2013;2:635–88.
11. Tang J, Aittokallio T. Network pharmacology strategies toward 31. Zhu H. Big data and artificial intelligence modeling for drug
multi-target anticancer therapies: from computational models discovery. Annu Rev Pharmacol Toxicol 2020;60:573–89.
to experimental design principles. Curr Pharm Des 2014;20: 32. Tripathi MK, Nath A, Singh TP, et al. Evolving scenario of big
23–36. data and artificial intelligence (AI) in drug discovery. Mol Divers
12. Hopkins AL. Network pharmacology. Nat Biotechnol 2007;25: 2021;25:1439–60.
1110–1. 33. Díaz Ó, Dalton JAR, Giraldo J. Artificial intelligence: a novel
13. Noor F, Tahir Ul Qamar M, Ashfaq UA, et al. Network pharma- approach for drug discovery. Trends Pharmacol Sci 2019;40:550–1.
cology approach for medicinal plants: review and assessment. 34. Raghupathi W, Raghupathi V. Big data analytics in healthcare:
Pharmaceuticals 2022;15:1–33. promise and potential. Health Inf Sci Syst 2014;2:3.
14. Hopkins AL. Network pharmacology: the next paradigm in drug 35. Sagiroglu S, Sinanc D. Big data: a review. In: International Con-
discovery. Nat Chem Biol 2008;4:682–90. ference on Collaboration Technologies and Systems (CTS). San Diego,
15. Chandran U, Mehendale N, Patil S, et al. Network Pharmacology. CA, USA: IEEE, 2013, 42–7.
In Innovative Approaches in Drug Discovery. Amsterdam: Elsevier, 36. Gandomi A, Haider M. Beyond the hype: big data concepts,
2017, 127. methods, and analytics. Int J Inf Manage 2015;35:137–44.
16. Batool S, Javed MR, Aslam S, et al. Network pharmacol- 37. Tripathi R, Sharma P, Chakraborty P, et al. Next-generation
ogy and bioinformatics approach reveals the multi-target sequencing revolution through big data analytics. Front Life Sci
pharmacological mechanism of Fumaria indica in the treat- 2016;9:119–49.
ment of liver cancer. Pharmaceuticals 2022;15:654. https://doi. 38. Pal S, Mondal S, Das G, et al. Big data in biology: the hope and
org/10.3390/ph15060654. present-day challenges in it. Gene Rep 2020;21:100869.
17. Zimmermann GR, Lehár J, Keith CT. Multi-target therapeutics: 39. Guoqing Z, Yixue L, Zefeng W, et al. New challenges and
when the whole is greater than the sum of the parts. Drug trends in bio-med big data. Bull. Chin. Acad. Sci. (Chinese Version)
Discov Today 2007;12:34–42. 2018;33:853–60.
18. Li S, Zhang B. Traditional Chinese medicine network pharma- 40. Lo Y-C, Ren G, Honda H, et al. Artificial intelligence-based drug
cology: theory, methodology and application. Chin J Nat Med design and discovery. In: Rasul A, Hussain G (eds.). Cheminfor-
2013;11:110–20. matics and Its Applications. New York, NY: IntechOpen, 2019.
19. Yuan H, Ma Q, Cui H, et al. How can synergism of traditional 41. Guo F, Tang X, Zhang W, et al. Exploration of the mechanism
medicines benefit from network pharmacology? Molecules of traditional Chinese medicine by AI approach using unsu-
2017;22:1135–54. pervised machine learning for cellular functional similarity of
20. Niu WH, Wu F, Cao WY, et al. Network pharmacology compounds in heterogeneous networks, XiaoErFuPi granules
for the identification of phytochemicals in traditional Chi- as an example. Pharmacol Res 2020;160:105077.
nese medicine for COVID-19 that may regulate interleukin- 42. Lehár J, Krueger AS, Avery W, et al. Synergistic drug combina-
6. Biosci Rep 2021;41:BSR20202583. https://doi.org/10.1042/ tions tend to improve therapeutically relevant selectivity. Nat
BSR20202583. Biotechnol 2009;27:659–66.
21. Gao X, Li S, Cong C, et al. A network pharmacology approach 43. Li TH, Wang CC, Zhang L, Chen X. SNRMPACDC: computa-
to estimate potential targets of the active ingredients of tional model focused on Siamese network and random matrix
Machine learning for synergistic network pharmacology | 23

projection for anticancer synergistic drug combination predic- 63. Cheng B-C, Tsai Y-A, Liao G-T, et al. HMM machine learning and
tion. Brief Bioinform 2023;24:bbac503. https://doi.org/10.1093/ inference for activities of daily living recognition. J Supercomput
bib/bbac503. 2010;54:29–42.
44. Chen X, Ren B, Chen M, et al. NLLSS: predicting synergistic drug 64. Noble WS. What is a Support Vector Machine? Nat Biotechnol
combinations based on semi-supervised learning. PLoS Comput 2006;24:1565–7.

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
Biol 2016;12:e1004975. 65. Ukil A, Ukil A. Support Vector Machine. Intell Syst Signal Process
45. Wang Z, Jensen MA, Zenklusen JC. A practical guide to The Power Eng 2007;161–226.
Cancer Genome Atlas (TCGA). Methods Mol Biol (Clifton, NJ) 66. Tsiligaridis J. Classification with neural network and SVM via
2016;1418:111–41. decision tree algorithm. In: AIP Conference Proceedings, 2018, p.
46. Parkinson H, Kapushesky M, Shojatalab M, et al. ArrayExpress– 020040. New York, USA: AIP Publishing LLC.
a public database of microarray experiments and gene expres- 67. Bhavsar H, Panchal MH. A review on Support Vector Machine
sion profiles. Nucleic Acids Res 2007;35:D747–50. for data classification. Int J Adv Res Comput Eng Technol 2012;1:
47. Edgar R, Domrachev M, Lash AE. Gene expression omnibus: 185–9.
NCBI gene expression and hybridization array data repository. 68. Dai W, Chen HY, Chen CY. A network pharmacology-based
Nucleic Acids Res 2002;30:207–10. approach to investigate the novel TCM formula against Hunt-
48. van IJzendoorn DGP, Szuhai K, Briaire-de Bruijn IH, et al. ington’s disease and validated by support vector machine
Machine learning analysis of gene expression data reveals model. Evid Based Complementary Altern Med 2018;2018:6020197.
novel diagnostic and prognostic biomarkers and identifies 69. Zernov VV, Balakin KV, Ivaschenko AA, et al. Drug discovery
therapeutic targets for soft tissue sarcomas. PLoS Comput Biol using Support Vector Machines. The case studies of drug-
2019;15:e1006826. likeness, agrochemical-likeness, and enzyme inhibition predic-
49. Baranwal M, Magner A, Elvati P, et al. A deep learning architec- tions. J Chem Inf Model 2003;43:2048–56.
ture for metabolic pathway prediction. Bioinformatics 2020;36: 70. Lei T, Li Y, Song Y, et al. ADMET evaluation in drug discovery: 15.
2547–53. Accurate prediction of rat oral acute toxicity using relevance
50. Monteiro NR, Ribeiro B, Arrais JP. Deep Neural Network archi- vector machine and consensus modeling. J Chem 2016;8:6.
tecture for drug-target interaction prediction. In: International 71. Boateng EY, Otoo J, Abaye DA. Basic tenets of classification
Conference on Artificial Neural Networks 2019, Munuch, Germany, algorithms K-Nearest-Neighbor, Support Vector Machine, Ran-
2019, p. 804–9. Cham, Germany: Springer. dom Forest and neural network: a review. J Data Anal Inform
51. El-Behery H, Attia AF, El-Feshawy N, et al. Efficient machine Process 2020;8:341–57.
learning model for predicting drug-target interactions with 72. Dara S, Dhamercherla S, Jadav SS, et al. Machine learning in
case study for Covid-19. Comput Biol Chem 2021;93:107536. drug discovery: a review. Artif Intell Rev 2022;55:1947–99.
52. Zong N, Li N, Wen A, et al. BETA: a comprehensive bench- 73. Degenhardt F, Seifert S, Szymczak S. Evaluation of variable
mark for computational drug-target prediction. Brief Bioinform selection methods for Random Forests and omics data sets.
2022;23:bbac199. https://doi.org/10.1093/bib/bbac199. Brief Bioinform 2019;20:492–503.
53. Huang K, Fu T, Glass LM, et al. DeepPurpose: a deep learn- 74. Chen HY, Chen JQ, Li JY, et al. Deep learning and Random Forest
ing library for drug-target interaction prediction. Bioinformatics approach for finding the optimal traditional Chinese medicine
2021;36:5545–7. formula for treatment of Alzheimer’s disease. J Chem Inf Model
54. Wan F, Hong L, Xiao A, et al. NeoDTI: neural integration of 2019;59:1605–23.
neighbor information from a heterogeneous network for dis- 75. Wu L, Gao X, Wang L, et al. Prediction of multi-target of
covering new drug-target interactions. Bioinformatics 2019;35: Aconiti Lateralis radix Praeparata and its network pharmacol-
104–11. ogy. Zhongguo Zhong Yao Za Zhi 2011;36:2907–10.
55. Surden H. Machine learning and law. Wash L Rev 2014;89:87. 76. Chen ZH, You ZH, Guo ZH, et al. Prediction of drug-target inter-
56. Lavecchia A. Machine-learning approaches in drug discovery: actions from multi-molecular network based on deep walk
methods and applications. Drug Discov Today 2015;20:318–31. embedding model. Front Bioeng Biotechnol 2020;8:338.
57. Lima AN, Philot EA, Trossini GH, et al. Use of machine learning 77. Naorem LD, Muthaiyan M, Venkatesan A. Integrated network
approaches for novel drug discovery. Expert Opin Drug Deliv analysis and machine learning approach for the identification
2016;11:225–39. of key genes of triple-negative breast cancer. J Cell Biochem
58. Lo YC, Rensi SE, Torng W, et al. Machine learning in chemoin- 2019;120:6154–67.
formatics and drug discovery. Drug Discov Today 2018;23: 78. Deng L, Yu D. Deep learning: methods and applications. Found
1538–46. Trends Signal Process 2014;7:197–387.
59. Baldi P. Autoencoders, unsupervised learning, and deep archi- 79. Bengio Y, Lamblin P, Popovici D, et al. Greedy layer-wise training
tectures. In: Proceedings of ICML workshop on unsupervised and of deep networks. Adv Neural Inf Process Syst 2006;19:153–60.
transfer learning, 2012, p. 37–49. JMLR Workshop and Conference 80. Pal SK, Mitra S. Multilayer Perceptron, fuzzy sets, and classifi-
Proceedings. cation. IEEE Trans Neural Netw 1992;3:683–97.
60. Han K, Wang Y, Zhang C et al. Autoencoder inspired unsu- 81. Stokes A, Hum W, Zaslavsky J. A minimal-input Multi-
pervised feature selection. In: IEEE International Conference on layer Perceptron for predicting drug-drug interactions with-
Acoustics, Speech and Signal Processing (ICASSP), 2018, p. 2941–5. out knowledge of drug structure. STEM Fellowsh J 2020;6:
Calgary, Alberta, Canada: IEEE. 19–23.
61. Hu Q, Feng M, Lai L, et al. Prediction of drug-likeness using deep 82. Bustamam A, Yanuar A, Mangunwardoyo W, et al. Comparison
autoencoder neural networks. Front Genet 2018;9:585. accuracy of Multi-Layer Perceptron and DNN in QSAR clas-
62. Emdadi A, Eslahchi C. Auto-HMM-LMF: feature selection based sification for acetylcholinesterase inhibitors. In: 2021 Interna-
method for prediction of drug response via autoencoder and tional Conference on Artificial Intelligence and Mechatronics Systems
hidden Markov model. BMC Bioinform 2021;22:33. (AIMS), 2021, p. 1–6. Bandung, Indonesia: IEEE.
24 | Noor et al.

83. Altalib MK, Salim N. Similarity-based virtual screen using 103. Hanneke S. Theoretical Foundations of Active Learning. Pennsylva-
enhanced Siamese deep learning methods. ACS Omega 2022;7: nia, USA: Carnegie Mellon University, 2009.
4769–86. 104. Alzubaidi L, Zhang J, Humaidi AJ, et al. Review of deep learning:
84. Bengio Y. Learning deep architectures for AI. Found Trends Mach concepts, CNN architectures, challenges, applications, future
Learn 2009;2:1–127. directions. J Big Data 2021;8:53.

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
85. Ramsundar B, Liu B, Wu Z, et al. Is multitask deep learning 105. Mamoshina P, Vieira A, Putin E, et al. Applications of deep
practical for pharma? J Chem Inf Model 2017;57:2068–76. learning in biomedicine. Mol Pharm 2016;13:1445–54.
86. Grebner C, Matter H, Kofink D, et al. Application of deep neural 106. Thompson NC, Greenewald K, Lee K, et al. The computational
network models in drug discovery programs. ChemMedChem limits of deep learning. arXiv preprint arXiv:2007.05558. 2020.
2021;16:3772–86. 107. Cai C, Wang S, Xu Y, et al. Transfer learning for drug discovery.
87. Chen J, Cheong C, Lan L, et al. DeepDRIM: a Deep Neural Net- J Med Chem 2020;63:8683–94.
work to reconstruct cell-type-specific gene regulatory network 108. Elbadawi M, Gaisford S, Basit AW. Advanced machine-learning
using single-cell RNA-seq data. Brief Bioinform 2021;22:bbab325. techniques in drug discovery. Drug Discov Today 2021;26:769–77.
https://doi.org/10.1093/bib/bbab325. 109. Zhuang F, Qi Z, Duan K, et al. A comprehensive survey on
88. Valueva MV, Nagornov N, Lyakhov PA, et al. Application of transfer learning. Proc IEEE Inst Electr Electron Eng 2021;109:
the residue number system to reduce hardware costs of the 43–76.
Convolutional Neural Network implementation. Math Comput 110. Devlin J, Chang M-W, Lee K, et al. Bert: pre-training of deep
Simul 2020;177:232–43. bidirectional transformers for language understanding. arXiv
89. Venkatesan R, Li B. Convolutional Neural Networks in Visual Com- preprint arXiv:1810.04805. 2018.
puting: A Concise Guide. London, UK: CRC Press, 2017. 111. Peng Y, Yan S, Lu Z. Transfer learning in biomedical natural
90. Hamza H, Nasser M, Salim N, et al. Bioactivity prediction using language processing: an evaluation of BERT and ELMo on ten
Convolutional Neural Network. In: Emerging Trends in Intelligent benchmarking datasets. arXiv preprint arXiv:1906.05474. 2019.
Computing and Informatics: Data Science, Intelligent Information 112. Lee J, Yoon W, Kim S, et al. BioBERT: a pre-trained biomedical
Systems and Smart Computing 4, 2020, p. 341–51. Johor, Malaysia: language representation model for biomedical text mining.
Springer. Bioinformatics 2020;36:1234–40.
91. Dhruv P, Naskar S. Image Classification Using Convolutional Neural 113. Zhu Y, Li L, Lu H, et al. Extracting drug-drug interactions from
Network (CNN) and Recurrent Neural Network (RNN): a review. texts with BioBERT and multiple entity-aware attentions. J
Singapore: Springer, 2020, 367–81. Biomed Inform 2020;106:103451.
92. Dupond S. A thorough review on the current advance of neural 114. Aldahdooh J, Vähä-Koskela M, Tang J, et al. Using BERT to iden-
network structures. Annu Rev Control 2019;14:200–30. tify drug-target interactions from whole PubMed. BMC Bioinform
93. Nag S, Baidya ATK, Mandal A, et al. Deep learning tools 2022;23:245.
for advancing drug discovery and development. 3 Biotech 115. Zheng J, Xiao X, Qiu WR. DTI-BERT: identifying drug-target
2022;12:110. interactions in cellular networking based on BERT and deep
94. Camargo G, Bugatti PH, Saito PTM. Active semi-supervised learning method. Front Genet 2022;13:859188.
learning for biological data classification. PloS One 116. Nguyen G, Dlugolinsky S, Bobák M, et al. Machine learning and
2020;15:e0237428. deep learning frameworks and libraries for large-scale data
95. Xia Z, Wu LY, Zhou X, Wong STC. Semi-supervised drug-protein mining: a survey. Artif Intell Rev 2019;52:77–124.
interaction prediction from heterogeneous biological spaces. 117. Liermann V. Overview machine learning and deep learning
BMC Syst Biol 2010;4(Suppl 2):S6. frameworks. In: The Digital Journey of Banking and Insurance, Vol.
96. Chen X, Liu MX, Yan GY. Drug-target interaction prediction by III: Data Storage, Data Processing and Data Analysis, New York:
random walk on the heterogeneous network. Mol Biosyst 2012;8: Palgrave Macmillan, 2021, 187–224.
1970–8. 118. Bisong E. Building Machine Learning and Deep Learning Models on
97. Chu X, Lin Y, Wang Y, et al. Mlrda: a multi-task semi-supervised Google Cloud Platform. Berlin, Germany: Springer, 2019.
learning framework for drug-drug interaction prediction. In: 119. Kuhn M. Building predictive models in R using the caret pack-
Proceedings of the 28th International Joint Conference on Artificial age. J Stat Softw 2008;28:1–26.
Intelligence, Macao, China, 2019, p. 4518–24. 120. Vasilev I, Slater D, Spacagna G, et al. Python Deep Learning: Explor-
98. Chen H, Zhang Z. A semi-supervised method for drug-target ing Deep Learning Techniques and Neural Network Architectures with
interaction prediction with consistency in networks. PloS One Pytorch, Keras, and TensorFlow. Birmingham, UK: Packt Publishing
2013;8:e62975. Ltd, 2019.
99. Yan C, Duan G, Zhang Y, et al. Predicting drug-drug interactions 121. Mishra P, CNN, RNN Using PyTorch. In PyTorch Recipes: A Problem-
based on integrated similarity and semi-supervised learning. Solution Approach to Build, Train and Deploy Neural Network Models.
IEEE/ACM Trans Comput Biol Bioinform 2022;19:168–79. Tampere, Finland: Springer, 2022, 49–115.
100. Song M, Yu H, Han WS. Combining active learning and semi- 122. Braun S. LSTM benchmarks for deep learning frameworks.
supervised learning techniques to extract protein interaction arXiv preprint arXiv:1806.01818. 2018.
sentences. Bioinformatics 2011;12(Suppl 12):S4. 123. Toleubay Y, James AP. Getting Started with TensorFlow Deep Learn-
101. Chowdhury MFM, Abacha AB, Lavelli A, et al. Two different ing. Cham: Springer, 2020.
machine learning techniques for drug-drug interaction extrac- 124. Parvat A, Chavan J, Kadam S et al. A survey of deep-learning
tion. In: Proceedings of DDI Extraction-2011 Challenge Task, Huelva, frameworks. In: 2017 International Conference on Inventive Systems
Spain, The 1st DDI Extraction-2011 challenge task, 2011, 761, p. and Control (ICISC), 2017, p. 1–7. Coimbatore, India: IEEE.
19–26. 125. Boss G, Malladi P, Quan D, et al. Cloud Computing, Vol. 321. New
102. Settles B. Active learning literature survey. Technical Report 1648. York, USA: IBM white paper, 2007, 224–31.
Wisconsin, WI: Department of Computer Sciences, University 126. Zaharia M, Xin RS, Wendell P, et al. Apache spark: a unified
of Wisconsin at Madison, 2009. engine for big data processing. Commun ACM 2016;59:56–65.
Machine learning for synergistic network pharmacology | 25

127. Pandey M, Fernandez M, Gentile F, et al. The transformational pharmacology. Chem Commun (Cambridge, England) 2019;55:
role of GPU computing and deep learning in drug discovery. Nat 6369–72.
Mach Intell 2022;4:211–21. 147. García-Campos MA, Espinal-Enríquez J, Hernández-Lemus E.
128. Mahmud M, Kaiser MS, McGinnity TM, et al. Deep learning in Pathway analysis: state of the art. Front Physiol 2015;6:383.
mining biological data. Cognit Comput 2021;13:1–33. 148. Jenwitheesuk E, Horst JA, Rivas KL, et al. Novel paradigms for

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
129. Mahmud M, Kaiser MS, Hussain A, et al. Applications of deep drug discovery: computational multitarget screening. Trends
learning and reinforcement learning to biological data. IEEE Pharmacol Sci 2008;29:62–71.
Trans Neural Netw Learn Syst 2018;29:2063–79. 149. Szklarczyk D, Gable AL, Nastou KC, et al. The STRING database
130. Reina GA, Gruzdev A, Foley P, et al. OpenFL: an open- in 2021: customizable protein-protein networks, and func-
source framework for federated learning. arXiv preprint tional characterization of user-uploaded gene/measurement
arXiv:2105.06413. 2021. sets. Nucleic Acids Res 2021;49:D605–d612.
131. Hauschild AC, Lemanczyk M, Matschinske J, et al. Federated 150. Szklarczyk D, Santos A, von Mering C, et al. STITCH 5: aug-
Random Forests can improve local performance of predic- menting protein-chemical interaction networks with tissue
tive models for various healthcare applications. Bioinformatics and affinity data. Nucleic Acids Res 2016;44:D380–4.
2022;38:2278–86. 151. Piñero J, Queralt-Rosinach N, Bravo À, et al. DisGeNET: a discov-
132. Rollinger JM, Stuppner H, Langer T. Virtual screening for ery platform for the dynamical exploration of human diseases
the discovery of bioactive natural products. Prog Drug Res and their genes. Database 2015;2015:bav028.
2008;65:211, 213–49. 152. Dale JM, Popescu L, Karp PD. Machine learning methods for
133. Zhi HY, Zhao L, Lee CC, Chen CYC. A novel graph neu- metabolic pathway prediction. BMC Bioinform 2010;11:15.
ral network methodology to investigate dihydroorotate dehy- 153. Dongardive J, Jamkhedkar S. Classification of metabolic path-
drogenase inhibitors in small cell lung cancer. Biomolecules ways using machine learning techniques. In: Innovations in
2021;11:477. https://doi.org/10.3390/biom11030477. Bio-Inspired Computing and Applications: Proceedings of the 11th
134. Jacek K, Hanna P, Anna M, et al. The log P parameter as a International Conference on Innovations in Bio-Inspired Computing
molecular descriptor in the computer-aided drug design–an and Applications (IBICA 2020) held during December 16-18, 2020 11,
overview. Comput Meth Sci Technol 2012;18:81–8. 2021, p. 97–110. Cham, Germany: Springer.
135. Di L, Artursson P, Avdeef A, et al. The critical role of pas- 154. Hao J, Kim Y, Kim TK, et al. PASNet: pathway-associated sparse
sive permeability in designing successful drugs. ChemMedChem Deep Neural Network for prognosis prediction from high-
2020;15:1862–74. throughput data. BMC Bioinform 2018;19:510.
136. Hashemifar S, Neyshabur B, Khan AA, et al. Predicting protein- 155. Rost B, Liu J, Nair R, et al. Automatic prediction of protein
protein interactions through sequence-based deep learning. function. Cell Mol Life Sci 2003;60:2637–50.
Bioinformatics 2018;34:i802–10. 156. Browne F, Zheng H, Wang H, Azuaje F. From experimen-
137. Lusci A, Pollastri G, Baldi P. Deep architectures and deep tal approaches to computational techniques: a review on
learning in chemoinformatics: the prediction of aqueous sol- the prediction of protein-protein interactions. Adv Artif Intell
ubility for drug-like molecules. J Chem Inf Model 2013;53: 2010;2010:1–15.
1563–75. 157. Hu X, Feng C, Ling T, et al. Deep learning frameworks for
138. Kucukdereli H, Allen NJ, Lee AT, et al. Control of excitatory protein-protein interaction prediction. Comput Struct Biotechnol
CNS synaptogenesis by astrocyte-secreted proteins Hevin and J 2022;20:3223–33.
SPARC. Proc Natl Acad Sci USA 2011;108:E440–9. 158. Du X, Sun S, Hu C, et al. DeepPPI: boosting prediction of protein-
139. Williams T, Tkachenko V. ChemSpider. Royal Society protein interactions with deep neural networks. J Chem Inf Model
of Chemistry, Cambridge. http://www.chemspider.com/. 2017;57:1499–510.
Accessed, 2016. 159. Chatterjee P, Basu S, Kundu M, et al. PPI_SVM: prediction of
140. Ayati A, Falahati M, Irannejad H, et al. Synthesis, in vitro anti- protein-protein interactions using machine learning, domain-
fungal evaluation and in silico study of 3-azolyl-4-chromanone domain affinities and frequency tables. Cell Mol Biol Lett
phenylhydrazones. Daru 2012;20:46. 2011;16:264–78.
141. Tetko IV, Gasteiger J, Todeschini R, et al. Virtual computational 160. Chen KH, Wang TF, Hu YJ. Protein-protein interaction pre-
chemistry laboratory–design and description. J Comput Aided diction using a hybrid feature representation and a stacked
Mol Des 2005;19:453–63. generalization scheme. BMC Bioinform 2019;20:308.
142. Manoj VM, Anunanthini P, Swathik PC, et al. Comparative anal- 161. Berger SI, Iyengar R. Network analyses in systems pharmacol-
ysis of glyoxalase pathway genes in Erianthus arundinaceus ogy. Bioinformatics 2009;25:2466–72.
and commercial sugarcane hybrid under salinity and drought 162. You Y, Lai X, Pan Y, et al. Artificial intelligence in cancer target
conditions. BMC Genomics 2019;19:986. identification and drug discovery. Signal Transduct Target Ther
143. Zhang R, Zhu X, Bai H, et al. Network pharmacology databases 2022;7:156.
for traditional Chinese medicine: review and assessment. Front 163. Wu Y, Zhu Y, Xie N, et al. A network pharmacology approach to
Pharmacol 2019;10:123. explore active compounds and pharmacological mechanisms
144. Cantini L, Medico E, Fortunato S, et al. Detection of gene of a patented Chinese herbal medicine in the treatment of
communities in multi-networks reveals cancer drivers. Sci Rep endometriosis. PloS One 2022;17:e0263614.
2015;5:17386. 164. Zeng X, Zhu S, Lu W, et al. Target identification among known
145. Rodrigues T, Werner M, Roth J, et al. Machine intelligence drugs by deep learning from heterogeneous networks. Chem Sci
decrypts β-lapachone as an allosteric 5-lipoxygenase inhibitor. 2020;11:1775–97.
Chem Sci 2018;9:6899–903. 165. Zheng Y, Wu Z. A machine learning-based biological drug-
146. Rodrigues T, de Almeida BP, Barbosa-Morais NL, et al. Dis- target interaction prediction method for a tripartite heteroge-
secting celastrol with machine learning to unveil dark neous network. ACS Omega 2021;6:3037–45.
26 | Noor et al.

166. Song S, Zhou J, Li Y, et al. Network pharmacology and experi- discovery and development. Expert Opin Drug Deliv 2020;15:
mental verification based research into the effect and mech- 1165–80.
anism of Aucklandiae Radix-Amomi Fructus against gastric 187. Kwon Y, Shin WH, Ko J, Lee J. AK-Score: accurate protein-
cancer. Sci Rep 2022;12:9401. ligand binding affinity prediction using an ensemble of 3D-
167. Liu Y, Gu HY, Zhu J, et al. Identification of hub genes and convolutional neural networks. Int J Mol Sci 2020;21:8424.

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
key pathways associated with bipolar disorder based on https://doi.org/10.3390/ijms21228424.
weighted gene co-expression network analysis. Front Physiol 188. Tahir Ul Qamar M, Zhu XT, Chen LL, et al. Target-specific
2019;10:1081. machine learning scoring function improved structure-based
168. Liu G, Luo S, Lei Y, et al. A nine-hub-gene signature of metabolic virtual screening performance for SARS-CoV-2 drugs develop-
syndrome identified using machine learning algorithms and ment. Int J Mol Sci 2022;23:11003.
integrated bioinformatics. Bioengineered 2021;12:5727–38. 189. Hsin KY, Ghosh S, Kitano H. Combining machine learning
169. Li J, Zeng Z, Jiang X, et al. Stromal microenvironment pro- systems and multiple docking simulation packages to improve
moted infiltration in esophageal adenocarcinoma and squa- docking prediction reliability for network pharmacology. PloS
mous cell carcinoma: a multi-cohort gene-based analysis. Sci One 2013;8:e83922.
Rep 2020;10:18589. 190. Rouillard AD, Hurle MR, Agarwal P. Systematic interrogation of
170. Attene-Ramos MS, Miller N, Huang R, et al. The Tox21 robotic diverse Omic data reveals interpretable, robust, and generaliz-
platform for the assessment of environmental chemicals–from able transcriptomic features of clinically successful therapeu-
vision to reality. Drug Discov Today 2013;18:716–23. tic targets. PLoS Comput Biol 2018;14:e1006142.
171. Patlewicz G, Jeliazkova N, Safford RJ, et al. An evaluation of 191. Bisaso KR, Anguzu GT, Karungi SA, et al. A survey of machine
the implementation of the Cramer classification scheme in the learning applications in HIV clinical research and care. Comput
Toxtree software. SAR QSAR Environ Res 2008;19:495–524. Biol Med 2017;91:366–71.
172. Cheng F, Li W, Zhou Y, et al. admetSAR: a comprehensive source 192. Xia X. Bioinformatics and drug discovery. Curr Top Med Chem
and free tool for assessment of chemical ADMET properties. J 2017;17:1709–26.
Chem Inf Model 2012;52:3099–105. 193. Romano JD, Tatonetti NP. Informatics and computational
173. Lysenko A, Sharma A, Boroevich KA, et al. An integrative methods in natural product drug discovery: a review and per-
machine learning approach for prediction of toxicity-related spectives. Front Genet 2019;10:368.
drug safety. Life Sci Alliance 2018;1:e201800098. 194. Regan-Fendt KE, Xu J, DiVincenzo M, et al. Synergy from gene
174. Wang Z, Liang L, Yin Z, Lin J. Improving chemical similarity expression and network mining (SynGeNet) method predicts
ensemble approach in target prediction. J Chem 2016;8:20. synergistic drug combinations for diverse melanoma genomic
175. Pu L, Naderi M, Liu T, et al. eToxPred: a machine learning- subtypes. NPJ Syst Biol Appl 2019;5:6.
based approach to estimate the toxicity of drug candidates. 195. Ambure P, Halder AK, Gonzalez Diaz H, et al. QSAR-Co: an
BMC Pharmacol Toxicol 2019;20:2. open source software for developing robust multitasking or
176. Pires DE, Blundell TL, Ascher DB. pkCSM: predicting small- multitarget classification-based QSAR models. J Chem Inf Model
molecule pharmacokinetic and toxicity properties using graph- 2019;59:2538–44.
based signatures. J Med Chem 2015;58:4066–72. 196. Wang Y-L, Wang F, Shi X-X, et al. Cloud 3D-QSAR: a web
177. Cañada A, Capella-Gutierrez S, Rabal O, et al. LimTox: a web tool for the development of quantitative structure–activity
tool for applied text mining of adverse event and toxicity relationship models in drug discovery. Brief Bioinform 2021;22:
associations of compounds, drugs and genes. Nucleic Acids Res bbaa276.
2017;45:W484–w489. 197. Gupta R, Srivastava D, Sahu M, et al. Artificial intelligence to
178. Mayr A, Klambauer G, Unterthiner T, Hochreiter S. Deep- deep learning: machine intelligence approach for drug discov-
Tox: toxicity prediction using deep learning. Front Environ Sci ery. Mol Divers 2021;25:1315–60.
2016;3:80. 198. Martinez V, Navarro C, Cano C, et al. DrugNet: network-based
179. Gururaj H, Flammini F, Kumari HC, et al. Classification of drug–disease prioritization by integrating heterogeneous data.
drugs based on mechanism of action using machine learning Artif Intell Med 2015;63:41–9.
techniques. Discover Artif Intell 2021;1:1–14. 199. Wang Y-Y, Cui C, Qi L, et al. DrPOCS: drug repositioning based
180. Gao S, Han L, Luo D, et al. Modeling drug mechanism of action on projection onto convex sets. IEEE/ACM Trans Comput Biol
with large scale gene-expression profiles using GPAR, an artifi- Bioinforma 2018;16:154–62.
cial intelligence platform. Bioinformatics 2021;22:17. 200. Capuzzi SJ, Kim IS-J, Lam WI, et al. Chembench: a publicly
181. Liu C, Hogan AM, Sturm H, et al. Deep learning-driven predic- accessible, integrated cheminformatics portal. J Chem Inf Model
tion of drug mechanism of action from large-scale chemical- 2017;57:105–8.
genetic interaction profiles. J Chem 2022;14:12. 201. Pires DE, Blundell TL, Ascher DB. mCSM-lig: quantifying the
182. Fronsdal T, Fronsdal K. Deep learning approaches for predicting effects of mutations on protein-small molecule affinity in
drug mechanisms of action (healthcare). genetic disease and emergence of drug resistance. Sci Rep
183. Jumper J, Evans R, Pritzel A, et al. Highly accurate pro- 2016;6:1–8.
tein structure prediction with AlphaFold. Nature 2021;596: 202. Pires DE, Ascher DB. CSM-lig: a web server for assessing and
583–9. comparing protein–small molecule affinities. Nucleic Acids Res
184. Baek M, Baker D. Deep learning and protein structure modeling. 2016;44:W557–61.
Nat Methods 2022;19:13–4. 203. Pires DE, Ascher DB. mCSM-AB: a web server for predicting
185. Carpenter KA, Huang X. Machine learning-based virtual antibody–antigen affinity changes upon mutation with graph-
screening and its applications to Alzheimer’s drug discovery: based signatures. Nucleic Acids Res 2016;44:W469–73.
a review. Curr Pharm Des 2018;24:3347–58. 204. Kaminskas LM, Pires DE, Ascher DB. dendPoint: a web resource
186. Serafim MSM, Kronenberger T, Oliveira PR, et al. The application for dendrimer pharmacokinetics investigation and prediction.
of machine learning techniques to innovative antibacterial Sci Rep 2019;9:1–9.
Machine learning for synergistic network pharmacology | 27

205. Montanari F, Knasmüller B, Kohlbacher S, et al. Vienna liver- 211. Xuan P, Cui H, Shen T, et al. Heterodualnet: a dual Convo-
tox workspace—a set of machine learning models for predic- lutional Neural Network with heterogeneous layers for drug-
tion of interactions profiles of small molecules with trans- disease association prediction via chou’s five-step rule. Front
porters relevant for regulatory agencies. Front Chem 2020;7: Pharmacol 2019;10:1301.
899. 212. Jiang H-J, Huang Y-A, You Z-H. SAEROF: an ensemble approach

Downloaded from https://academic.oup.com/bib/article/24/3/bbad120/7109956 by Chulalongkorn University, Center for Academic Reso user on 01 December 2023
206. Fahimian G, Zahiri J, Arab SS, Sajedi RH. RepCOOL: computa- for large-scale drug-disease association prediction by incor-
tional drug repositioning via integrating heterogeneous biolog- porating rotation forest and sparse autoencoder Deep Neural
ical networks. J Transl Med 2020;18:1–10. Network. Sci Rep 2020;10:1–11.
207. Wang M-N, You Z-H, Li L-P et al. WGMFDDA: a novel weighted- 213. Lee I, Keum J, Nam H. DeepConv-DTI: prediction of drug-target
based graph regularized matrix factorization for predicting interactions via deep learning with convolution on protein
drug-disease associations. In: International Conference on Intelli- sequences. PLoS Comput Biol 2019;15:e1007129.
gent Computing, 2020, p. 542–51. Switzerland: Springer. 214. Abdel-Basset M, Hawash H, Elhoseny M, et al. DeepH-DTA: deep
208. Banerjee P, Eckert AO, Schrey AK, et al. ProTox-II: a webserver learning for predicting drug-target interactions: a case study
for the prediction of toxicity of chemicals. Nucleic Acids Res of COVID-19 drug repurposing. IEEE Access 2020;8:170433–51.
2018;46:W257–63. https://doi.org/10.1109/access.2020.3024238.
209. Dong J, Wang N-N, Yao Z-J, et al. ADMETlab: a platform for 215. Shen C, Luo J, Ouyang W, et al. IDDkin: network-based influ-
systematic ADMET evaluation based on a comprehensively ence deep diffusion model for enhancing prediction of kinase
collected ADMET database. J Chem 2018;10:1–11. inhibitors. Bioinformatics 2020;36:5481–91.
210. Oldenhof M, Arany A, Moreau Y, et al. ChemGrapher: optical 216. Hornig M, Klamt A. COSMO f rag: a novel tool for high-
graph recognition of chemical compounds by deep learning. J throughput ADME property prediction and similarity screening
Chem Inf Model 2020;60:4506–17. based on quantum chemistry. J Chem Inf Model 2005;45:1169–77.

You might also like