0% found this document useful (0 votes)
83 views31 pages

Artificial Intelligence For Drug Toxicity and Safety: Columbia University Medical Center New York, NY

This document discusses using artificial intelligence and machine learning to improve drug safety assessment. It describes how AI can help address limitations in traditional pre-clinical and post-marketing drug safety evaluation tools. Specifically, it explores how machine learning and deep learning have been applied to pre-clinical toxicity prediction using quantitative structure-activity relationship models and how AI is being used to analyze post-marketing adverse event reports to enhance pharmacovigilance.

Uploaded by

Arman
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
83 views31 pages

Artificial Intelligence For Drug Toxicity and Safety: Columbia University Medical Center New York, NY

This document discusses using artificial intelligence and machine learning to improve drug safety assessment. It describes how AI can help address limitations in traditional pre-clinical and post-marketing drug safety evaluation tools. Specifically, it explores how machine learning and deep learning have been applied to pre-clinical toxicity prediction using quantitative structure-activity relationship models and how AI is being used to analyze post-marketing adverse event reports to enhance pharmacovigilance.

Uploaded by

Arman
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 31

Artificial

Intelligence for Drug Toxicity and Safety


Anna O. Basilea,*, Alexandre Yahia,*, Nicholas P. Tatonettia,1

aColumbia University Medical Center New York, NY

*Contributed equally to this work


1Corresponding author

Email address: [email protected] (Nicholas P. Tatonetti)

Abstract
Interventional pharmacology is one of medicine’s most potent weapons
against disease. These drugs, however, can result in damaging side effects
and must be closely monitored. Pharmacovigilance is the field of science that
monitors, detects, and prevents adverse drug effects. Safety efforts begin
during the development process, using in vivo and in vitro studies, continue
through clinical trials, and extend to post-marketing surveillance of ADRs in
real-world populations. Future toxicity and safety challenges, including
increased polypharmacy and patient diversity, stress the limits of these
traditional tools. Massive amounts of newly available data present an
opportunity for using artificial intelligence and machine learning to improve
drug safety science. Here, we explore recent advances as applied to pre-
clinical drug safety and post-marketing surveillance with a specific focus on
machine and deep learning approaches.
Keywords:
Pharmacovigilance, Machine Learning, Deep Learning, Adverse Drug
Reactions

The challenge of keeping drugs safe
Drug safety is a major challenge in bring new drugs to market. Unexpected
toxicities are a major source of attrition during clinical trials and post-


marketing safety concerns cause unnecessary morbidity and mortality.
Adverse events (AEs), or adverse drug reactions (ADRs) when causality is
demonstrated, are unexpected effects occurring from a normal dosage of the
drug. Between 2008 and 2017, the Food and Drug Administration (FDA)
approved 321 novel drugsi. Over the same period of time, the FDA Adverse
Event Reporting System (FAERS) ii recorded more than 10 million AE reports,
among which 5.8 million were serious reports and 1.1 million were AEs
related to death. AEs burden our health system causing 2 million hospital
stays each year and lengthening visits by 1.7 to 4.6 daysi. The economic,
social, and health burden of toxicity and safety assessment is an essential and
pressing public health concern.
There are two complementary systems to address drug safety (Figure 1).
Before a drug is approved, clinical trials ensure that this drug is safe and
effective for its intended use. Once a drug is marketed, drugs are monitored
through AE reports to ensure a drug’s safety information is up to date, a
process called pharmacovigilance (PV). However, neither of these processes
are error proof as clinical trials suffer from structural limitations. For
example, it is impossible to test for all potential synergistic effects or to
conduct trials on populations large enough to detect rare AEs. Until recently,
women and the elderly were considered special sub-groups for clinical trials.
These trials have focused on designing drugs for the average patient [1] even
at a time when there are increasing calls for precision medicine to enable the
“right drug at the right dose to the right patient”[2]. Once drugs are approved,
it is the purview of programs to monitor drug safety. These agencies use
databases of spontaneously collected AE reports to flag leads and perform
confirmatory follow up analyses. However, these spontaneous reports are
known to suffer from biases such as under-reporting which is especially
troublesome for rare events and drug-drug interactions (DDIs)[3]. The
research community has turned to statistical and computational approaches
to address these limitations and supplement its PV toolbox [4, 5].
Over the past decade, we have seen two phenomena occur: (1) the
explosion of freely accessible databases of medical, chemical, and
pharmacological knowledge, along with the rapid adoption of electronic
health record (EHR) systems stimulated by the Health Information
Technology for Economic and Clinical Health (HITECH) Actiii; and (2) the
development of novel computational methods in the realm of machine
learning (ML) (see Glossary) and deep learning (DL) a popular re-branding
of neural networks - catalyzed by exponential increase in compute power and

2
data availability. Below, we explore the recent literature leveraging artificial
intelligence (AI) methods, both ML and DL, on novel data sources for pre-
clinical and in post-marketing surveillance for PV (Figure 1). We further
encourage the reader to reference the following introductory reviews for
more details on ML [6, 7, 8] and DL[9, 10].

Pre-clinical Drug Safety


AI techniques have been shown to play an important role in pre-market
drug safety, especially in the area of toxicity evaluation. Drug toxicity
determination is a main step in drug design and involves identifying the AEs
of chemicals on humans, plants, animals, and the environment[11]. Pre-
clinical evaluations are a necessity for preventing toxic drugs from reaching
clinical trials. Despite this, high toxicity is still a major contributor to drug
failure accounting for two-thirds of post-market drug withdrawals[12] and
for one-fifth of failures during clinical trials[13]. Thus, accurate toxicity
estimates are necessary for ensuring drug safety, and can help reduce the
cost and development time of bringing new drugs to market. Animal studies
have historically been the most conventional approach taken to assess
toxicity[11, 12, 13]. However, these studies are constrained by cost, time, and
ethical considerations. Numerous computational, in silico, approaches have
demonstrated utility in estimating the toxicity of drug candidates. These
approaches predict toxicity by evaluating various features of the drug and
include target-based predictions and Quantitative Structure-Activity
Relationships (QSAR). Below, we focus largely on ML and DL approaches in
the area of QSAR and discuss DL techniques in toxicity prediction and
assessment.

Quantitative Structure-Activity Relationships (QSAR)


QSAR is a method that establishes quantitative relationships between
chemical or structural characteristics and pharmacological activity[14].
QSAR methods have been used to model numerous drug safety endpoints
including drug lethal dose 50% (LD50)values, skin/eye irritation, and
tissue-specific toxicity[15]. Specifically, a QSAR model can analyze the
relationship between several predictors (e.g. molecular properties) and a
response (e.g. biological activities such as binding affinity)[16]. Good models
will be highly predictive and fairly easy to interpret. There are several types
of ML approaches that have been used for QSAR modeling.

3
Regression
Early QSAR techniques relied on multivariate linear regression to assess
the chemical properties of drug candidates[17]. These approaches are
sensitive to high data dimensionality and feature correlation which may
result in overfitting, and limited interpretability. Modern regression-based
approaches incorporate feature selection techniques to address these
concerns. One such technique is the use of a penalized regression model. L1
regularization, which is used in least absolute shrinkage and selection
operator (LASSO), aims to prevent overfitting by reducing the number of
features and only selecting subsets that are most relevant to the QSAR
model prediction[18]. L2 regularization, which is used in ridge regression
(RR), aims to alleviate collinearity by reducing the effective number of
features used in the model. Recently, Algamal et al.[16] proposed a weight
adjustment to the adaptive LASSO aimed to improve the selection of
correlated descriptors. This approach demonstrated potential when used to
develop a QSAR prediction of the anti-cancer potency of various
imadazo[4,5-b]pyridine derivatives. The authors also proposed applying L1-
norm regularization in the selection of significant descriptors for anti-
hepatitis C virus activity of thiourea derivatives[19]. While regression-based
approaches have demonstrated utility in QSAR prediction, assumptions of
linearity, which are inherent in regression, as well as issues of
dimensionality affect most QSAR modelling tasks. Currently, the most
common alternatives are support vector machines (SVM) and ensemble
approaches, such as random forest, due to their high predictive accuracy,
robustness, and ease of interpretation[20].

Support Vector Machines


Support Vector Machines (SVM) is an approach that aims to find a
hyperplane in an n dimensional space (n is defined by the number of
features) that discriminatively classifies the data. For example, if there are
2 input features, the hyperplane is a line. With 3 features, the hyperplane
is a 2-dimensional plane[21]. Support vectors are data points used to
build the SVM. These data points are located near the hyperplane and
influence the orientation and the position of this hyperplane. These
support vectors are used to maximize the margin of the classifier. SVM
performs classification by using a kernel function to map vectors into a
higher dimensional feature space.

In a recent QSAR modeling of histone deacetylase 1 (HDAC1)


inhibitors, SVM exhibited the best performance in predicting activity value

4
when compared with naive Bayes, k-nearest neighbor (k-NN), and
random forest algorithms[22]. Specifically, when SVM was used in
conjunction with a Chemistry Development Kit (CDK)iv[23] molecular
fingerprint for HDAC1 activity prediction with 5-fold cross validation of
a training set, it achieved an area under the receiver operating curve
(AUC)=0.91, with 97% sensitivity, and 50% specificity. The training set
consisted of over 2,300 human HDAC1 inhibitors extracted from the
Binding Databasev, BindingDB[24]. This model also performed well
(AUC=0.89, with 95% sensitivity, and 75% specificity) when validated
using an external set of 413 ChEMBLvi compounds[25].

Another recent study by Nehoei et al.[26] used a genetic algorithm (GA)


[23] variable selection approach with SVM to develop QSAR models for the
prediction of vascular endothelial growth factor receptor 2 (VEGFR-2)
inhibition by aminopyrimidine-5-carbaldehyde oxime derivatives. More
recently, Algamal et al.[27] applied an L1-norm SVM approach to build a
QSAR classification model for neuraminidase inhibitors of influenza A virus
(H1N1).

Ensemble learning
Ensemble methods combine several ML models into a robust predictive
model. In doing so, they have improved predictive performance when
compared with a single model, and are often less susceptible to bias and
overfitting. Random forest is an ensemble learning algorithm that can handle
class imbalances and avoid overfitting, two common challenges in QSAR
modeling. Ng et al.[28] used a decision forest model to predict estrogen
receptor binding, using a 3,308 chemical training set from the FDA’s
Estrogenic Activity Databasevii. Models showed good performance with an
internal accuracy of 92% and an external validation ranging from 70-89%
accuracy. More recently, Lee et al.[29] demonstrated the use of random forest
in ligand-based QSAR modelling using ChEMBL bioactivity data. Training of
the 1,121 developed QSAR models showed an overall AUC = 0.97 using 5-fold
cross validation. Testing on an external validation set showed an accuracy of
0.89.
To further highlight the impact of class imbalances in QSAR modeling,
Grenet et al.[30] showed that many commonly used ML modeling approaches
under-performed when using data from ToxCastviii, an US Environmental
Protection Agency (EPA) managed public dataset containing chemical
structure and bioactivity data. These data are imbalanced, and highly

5
enriched for inactive compounds that are negative for toxicity in in vitro
assays and for whom a half maximal activity concentration (AC50) could
not be measured. When SVM, random forest, linear discriminant analysis
(LDA), and neural networks were used on the ToxCast data, the AUC ranged
between 0.6 and 0.73 across the methods, well below the expected
performance. In response, the authors developed a stacked generalization
approach, an ensemble method that consists of training a learning algorithm
by combining the predictions of other algorithms. This stacked approached
showed better performance with more models reaching an AUC>0.80 than
any of the single QSAR classifiers.

Software
QSAR can also be used to predict target-based activities like toxicity. The
field of target-driven toxicity prediction is heavily infiltrated with
proprietary tools. Many of these use classical ML algorithms but refine the
type of data used to calculate predictions. TargeTox[31] ix and PrOCTOR[32]
x are two examples of recent open-source toxicity prediction tools.

TargeTox leverages protein target data with a network-based approach


and gradient boosting to identify potentially toxic drugs. This approach
builds protein networks using a distance metric following the assumption
that neighboring biological entities share functional roles, thus hypothesizing
that toxicity responses can be isolated to specific network regions. The built
networks, pharmacological, and functional impact data from the public
datasets, like DrugBankxi[33] and ChEMBL[25] (Table 1), comprise the
model features. A gradient boosting classifier is then applied to develop a
quantitative toxicity prediction score for each drug. While the authors
specifically discuss a gradient boosting ensemble approach, any classifier
with a regularization function and the capacity to handle non-linear
relationships can be applied. TargeTox has multiple model variants based on
which distance metric is used for network calculation. The top performing
approach uses a diffusion state distance (DSD) with a subset of reference
points to calculate distance to the closest protein bound by a drug candidate.
This method achieved an AUC of .743 with a sensitivity of 0.75 and specificity
of 0.658 when trained and tested on data from DrugBank[33] and
ClinicalTrails.govxii with 5-fold cross validation. The novelty of TargeTox is its
ability to generate protein network data as well as combine other
pharmacological and functional features into a ML classifier for toxicity
prediction.

6
PrOCTOR is a target-based toxicity prediction software that in addition to
network information, also incorporates chemical properties into its scoring.
To develop a PrOCTOR score, the algorithm combines chemical structure
properties of the drug candidate (e.g. molecular weight, polar surface area,
quantitative estimate of drug-likeness (QED)) along with protein target
information (e.g. network connectivity, tissue-specific expression). Drug
target data is extracted from public datasets including DrugBank[33],
GTExxiii[34], and ExACxiv[35]. Compared to TargeTox, the PrOCTOR model
includes many more features with a total of 48 variables (34 target-based, 10
structure, and 4 drug-likeness) per drug compound model. A random forest
classifier is used on the 48-feature model to develop a PrOCTOR score which
assesses the likelihood of toxicity. This model constructs 50 decision trees
using a subset of the features and uses the tree consensus to predict outcome.
PrOCTOR showed high performance (AUC=0.83) and accuracy (ACC = 0.75)
with high sensitivity (0.75) and specificity (0.74) when trained on a set of
784 FDA drugs with 10-fold cross validation. Further, when tested on a set of
FDA-approved drugs, the algorithm scored 3 drugs with known toxicity
events, docetaxel, bortezomib, and rosiglitazone, with the worst score.
PrOCTOR’s ability to leverage multiple types of target and structure-based
features for toxicity prediction places sets it above many other target-based
algorithms.


Deep Learning
DL is an extension of ANNs which uses a hierarchy of ANNs to learn useful
features from raw data. A Merck-sponsored Kaggle competition in 2012
introduced DL to the field of drug discovery. The winning team used DL on a
set of diverse QSAR data sets to predict activity values for various
compounds[20]. Recently, numerous studies of toxicity modelling have used
DL approaches.
To assess hepatotoxicity, Xu et al. used DL to build drug-induced liver
injury (DILI) prediction models with chemical structure data[36]. The
authors used a recurrent neural network to construct the models. The best
model was trained on 475 drugs and predicted an external validation set of
about 200 drugs with an accuracy of 86.9%, sensitivity of 82.5%, and
specificity of 92.9%. This model outperformed previously reported DILI
prediction models.

7
Deep convolutional neural networks (CNNs) are a class of DL networks
that learn representations of raw images from pixel information as a
hierarchy of images from which features can be extracted and used to classify
complex patterns[37]. CNNs have been used to predict toxicity from images
of cells pre-treated with a set of drugs[37]. This approach was able to
effectively predict a broad spectrum of toxicity mechanisms from different
drugs, nuclear stains, and cell lines. Tong et al. also used a CNN strategy in a
protein structure analysis task[38]. Specifically, a 3D CNN approach was used
to analyze amino acid microenviroments and predict effects of mutations on
protein structure. No prior knowledge or feature assumptions were required
for this prediction task. And, the approach demonstrated a two-fold increase
in accuracy prediction compared with models that require hand-selected
features. Other DL approaches that have been used to assess drug toxicity
include autoencoders[39], generative adversarial networks (GANs)[40] and
long short-term memory (LSTM)[41], among others.

Post-marketing surveillance
In 1962, it was revealed that thousands of babies were born with
malformed limbs because thalidomide, a mild sleeping pill, had no
contraindications for pregnant women to whom it was often prescribed off-
label[42]. The WHO ”Programme for International Drug Monitoring”xv was
created following this disaster. Since 1978, the Uppsala Monitoring Centre
(UMC) in Sweden is the global coordinator for PV in collaboration with the
WHO, and counts 134 full member countries with national agencies
supporting patient safety and drug AE reporting systems. These initiatives
are the proof that safety assessment in clinical trials has its limit, and that
drug safety needs to be actively monitored during the entire market life of
drugs.
In the United States, the FDA maintains FAERS, a database containing
adverse event reports, medication error reports and product quality
complaints resulting in AEs. These self-reported Individual Case Safety
Reports (ICSRs) have been a major data source for post-marketing drug
safety mining. The classical methods to evaluate causality include the
Naranjo algorithm[43], the Venulet algorithm[44], and the World Health
Organization-Upsala Monitoring Centre (WHO-UMC) system for
standardized case causality assessment, among others[45].

8
Naturally, spontaneous reporting systems (SRS) like FAERS enabled data
mining methods to identify statistical associations between drugs and AEs
[46, 47]. But with known short-comings, such as confounding biases and
under-reporting[48], the attention has shifted to other data sources and
advanced computational methods that could replace or complement existing
resources. Below we discuss the approaches and data sources that can
support post-marketing PV along with the associated AI-driven methods
needed to extract information and learn from it.

System Pharmacology
System pharmacology is the study of drug action using principals from
systems biology, considering the effect of drug on the entire system rather
than a single target or metabolizing enzyme. This approach promises to
explain unexpected drug effects that may result from complex interactions of
targets and pathways. Application of systems pharmacology to adverse drug
events differs from its use in drug discovery in that it’s focused on off-target
effects and clinical observations of adverse reactions. In addition, it is one of
the most data-rich approach to drug safety for in silico ADR mining. A variety
of open databases are available and have been listed in Table 1. As a
consequence of the rich data sources available, investigators in system
pharmacology for adverse drug effects (ADEs) now have methods of choice
involving network approaches and the ability to integrate multiple types of
features. Lorberbaum et al.[57] proposed the modular assembly of drug
safety subnetworks (MADSS) where they generated protein networks using
knowledge bases that were pruned with literature mining, genome-wide
association study (GWAS) data, assigned phenotype target with DrugBank
and ChEMBL, and finally, trained random forest models on network metrics
to predict new drugs causing AEs. Raja et al.[58] focused on DDIs by mining
the literature to integrate drug-gene interactions (DGIs) at different scales
and trained random forest models to predict DDIs with a gold standard
corpus, focusing specifically on cutaneous diseases. Sornalakshmi et al.[59]
trained SVM models on similarity measures such as 2D molecular structure
similarity, 3D pharmacologic similarity, interaction profile fingerprint (IPF)
similarity, target similarity and ADE similarity from DrugBank and SIDERxvi
to predict drug pairs likely to interact with each other based on a literature-
based gold standard. Xu et al. [36] used various pharmaceutical compound
datasets and neural networks to encode these drugs using the undirected
graph recursive neural networks (UG-RNN) introduced by Lusci et al. [60]

9
and classified them between DILI positive and DILI negative compounds.
Herrero et al.[61] used pharmacokinetic (PK) and pharmacodynamic (PD)
properties from DrugBank and other sources, along with drug-enzymes
relationship data to build neural networks supervised models taking
Lexicompxvii and Vidal compendiaxviii as ground truth for DDI labels.

All of the above approaches heavily rely on molecular features directly


linked to these drugs along with phenotypic evidence of their effects, and the
source datasets are usually open. However, the clinical information used are
often limited to specific outcomes, missing on the longitudinal patient
medical history, and all the other clinical covariates. In contrast, EHRs are
closed datasets that have been used to compensate the episodic aspect of SRS
and provide observational data captured during medical encounters.

EHR mining
Stimulated by the HITECH Act, the rapid and widespread adoption of
EHRs that we have witnessed this past decade has also enabled researchers
to tap into these rich and noisy sources of clinical data for PV. EHR data stand
out in their challenging heterogeneity: these temporal data sources include
categorical data such as diagnostic, procedure and medication codes, but also
continuous laboratory tests and measurement values, along with large
volumes of semi-structured and unstructured medical notes and reports.

Structured EHR data


Structured data such as diagnoses, procedures, medications and
laboratory tests present the advantage of requiring the least pre-processing
for ML approaches. Zhao et al.[62] studied nine different weighting strategies
regarding how to use drugs, diagnoses and measurements as features in
supervised learning algorithms for ADR prediction.
Bayesian methods have been popular in modeling medical outcomes.
Benefiting from a knowledge-rich domain, a variety of Bayesian approach
have been used for adverse event prediction by including prior medical
knowledge. Bekker et al.[63] used Bayesian network representations to
model the effect of drugs on the progression of multiple co-morbidities using
prescriptions and diagnoses from primary care data. Moghaddass et al.[64]
proposed to generalize the self-controlled case series (SCCS) with a
multivariate hierarchical Bayesian model to leverage latent factor analysis
(LFA) and bring more interpretability regarding the effects of transient
multi-drug exposures on a collection of health outcomes. As an alternative,

10
Morel et al.[65] proposed another multivariate SCCS method based on
convolution of step functions with point drug exposures to estimate the effect
of longitudinal features. Kuang et al. directly followed up with that multiple
SCCS and presented a baseline regularization to take into account individual-
specific, time dependent occurrence rate of AEs. More recently, they have
also presented a version of that model for drug repurposing[66].

Rather than predicting discrete outcomes, it is also possible to model drug


responses by predicting dynamic time series of observational data to select
the best treatment courses. Xu et al. [67] estimated individualized treatment
response (ITR) curves with Bayesian non-parametrics (BNP). They
modeled creatinine time series response of treatments used in managing
kidney function and demonstrated a gain in accuracy compared to baseline
models. Structured data have the advantage of being easily pre-processed for
ML and DL algorithms, but they also have to rely on mappings, data structures
and terminologies that could impede reproducibility. More importantly,
because they are primarily designed for billing purposes, EHR databases
present a number of biases, including confounding bias and selection
bias[68], and do not provide the whole picture of their patients’ care
trajectory.

Clinical notes and biomedical corpora

Beyond structured data, which present both standardization and


mapping challenges and may not be readily accessible, biomedical and
clinical corpora represent a valuable resource. Clinical corpora enable the
learning of medical language model that embed prior knowledge and help
with the pragmatics of context-specific use of certain words. Natural
Language Processing (NLP) methods are essential in this area to extract
concepts and apply ML or learn embedding representations of these
documents to directly make predictions. Abacha et al.[69] developed a hybrid
feature-based and kernel-based system for DDI detection and classification
applied to the DDI-Extraction-2013 corpus. That corpus contained 1017
medical texts, including abstracts from MEDLINE and documents describing
DDIs from the DrugBank database, annotated with DDIs and pharmacological
substances for the supervision of the learning task. Mower et al.[70] built
embedding of semantic predications (ESP), by extracting concept-
relationship-concept triples from the literature with the SemRep NLP
systemxix. They trained a kNN model with the Exploring and Understanding

11
ADRs by Integrative Mining of Clinical Records and Biomedical Knowledge
(EU-ADR), and the Observational Medical Outcomes Partnership (OMOP)
datasets, two national networks that have defined common data models, as
ground truth and showed good generalization performances to predict
binary ADE outcomes. Kim et al.[71] opted for a naive Bayes classifier to
predict the likelihood of ADR in textual data from expert opinion on ADR case
reports from the Korean Adverse Event Reporting System database.
NLP has dramatically benefited from advances in DL in the recent years
to build better language models, with the development of word
embeddings[72], sequence to sequence (seq2seq) learning[73], and more
recently attention mechanisms[74, 75, 76]. The clinical domain has always
been a challenging field of application for NLP, and these novel methods have
been promptly applied to PV problems[77]. Language models can be trained
using the biomedical literature and then applied to clinical notes, as
demonstrated by Dev et al.[78], where the authors used MEDLINE to learn a
better representation of concepts found in narrative logs for classification of
ADEs. A recurrent theme in NLP for the detection of drug to AE relationship
prediction is the need to first detect the concept (i.e., Named Entity
Recognition, (NER)), and then perform the learning task. These two tasks as
showed in the studies previously mentioned can both be conducted with
neural networks. With its natural properties of connections and weights, DL
enables multi-task learning (MTL), an approach that consists of sharing the
weights of the neural networks between multiple tasks to improve overall
performances. Zhang et al.[79] used this technique to jointly learn NER in
texts for AE cases and ADE classification between serious and non-serious
effects. Similarly, Li et al. [80] applied biLSTM networks on the Medication,
Indication and Adverse Drug Events (MADE) 1.0 challenge for NER with a
conditional random field network, and for relations extraction with an
attention mechanism. More recently and using the same dataset, Yang et al.
[81] developed a similar LSTM model for NER but extracted relations
between concepts and ADEs by comparing SVM and random forests.
These NLP techniques have also been used with data collected on social
media and in online health communities. Some of the reviews covering
covering these applications are[82, 59, 83]. There is evidence that users on
social media disseminate information comparable to ICSRs and ADEs can be
classified from these high-noise data sources[84]. Post-marketing PV has
been conducted using Twitter data [85, 86] with embedding techniques and
biLSTM deep classifiers that outperform conditional random field methods,

12
discussion forums[87], or more domain-specific health social networking
sites[88].

Concluding Remarks
The availability of publicly accessible data, adoption of EHR systems, and
development of novel ML and DL approaches has transformed the field of PV.
This article focuses specifically on advances in AI techniques in the context of
pre-clinical drug safety and post-marketing surveillance. We encourage the
reader to reference systemic reviews on PV[21, 89, 90, 91, 92] which present
a more complete picture landscape of PV.
In the recent years, we have observed a growing integration of multi-scale
data, from molecular databases to clinical datasets, in conjunction with a
democratization of DL models to leverage these different data types. Neural
nets have been used so far mostly for NLP applications in PV, but they have
integrated the most recent state-of-the-art concepts such as attention
mechanisms and multi tasks learning. Their applications are starting to be
used beyond that scope, both in chemoinformatics and with clinical
observational data. We noted that most of the approaches in the recent years
that aim at predicting ADEs have been using annotated datasets. This almost
exclusive use of supervised models has its limits, as the prediction of novel
and unknown drug effects cannot rely on labeled data.
This is only the dawn of AI, and numerous questions remain such as how
to address class imbalances in supervised modeling tasks, and how to
incorporate unsupervised approaches in PV studies (Outstanding Questions).
Techniques such as GANs hold promise in addressing some of these concerns.
For example, novel unsupervised approaches using GANs that can generate in
silco molecules with desired chemical properties are starting to emerge[93,
94], showing great promise for drug safety.
While academic research has witnessed a drastic increase in the use of
ML and DL, the community will begin to see these approaches entering into
practice at a growing rate. For example, the FDA recently released plans for a
new regulatory framework to promote the development of safe medical
devices using AI algorithms. We expect that this will extend to drug
development and safety in the future. Appropriate regulatory frameworks
will need to be established to control for the risk of false positives. Overall,
the risk of implementing AI approaches for PV is low and the opportunity high
as it may have a positive impact on healthcare.

13

Conflict of interest: The authors do not have any conflicts of interest to


declare.

Acknowledgments: The authors are supported by NIH NIGMS


R01GM107145.

Resources

i) https://health.gov/

ii) https://open.fda.gov/data/faers/

iii) https://www.hhs.gov/hipaa/for-professionals/special-topics/ hitech-


act-enforcement-interim-final-rule/index.html

iv) https://cdk.github.io

v) https://www.bindingdb.org/bind/index.jsp

vi) https://www.ebi.ac.uk/chembl/

vii) https://www.fda.gov/science-research/bioinformatics-
tools/estrogenic-activity-database-eadb

viii) https://www.epa.gov/chemical-research/exploring-toxcast-data-
downloadable-data

ix) https://github.com/artem-lysenko/TargeTox

x) https://github.com/kgayvert/PrOCTOR

xi) https://www.drugbank.ca/

xii) https://clinicaltrials.gov/

14
xiii) https://gtexportal.org/home/

xiv) http://exac.broadinstitute.org/

xv) https://www.who.int/medicines/areas/quality_safety/safety_effi
cacy/National_PV_Centres_Map/en/

xvi) http://sideeffects.embl.de/

xvii) https://online.lexi.com

xviii) http://www.vidal-dis.com/

xix) https://semrep.nlm.nih.gov/

xx) https://www.ebi.ac.uk/chebi/

xxi) https://pubchem.ncbi.nlm.nih.gov/

xxii) https://reactome.org/

xxiii) https://www.genome.jp/kegg/


References

[1] C. Tannenbaum, D. Day, et al., Age and sex in drug development and
testing for adults, Pharmacological research 121 (2017) 83–93.

[2] F. S. Collins, H. Varmus, A new initiative on precision medicine, New


England Journal of Medicine 372 (9) (2015) 793–795.

[3] A. Marengoni, G. Onder, Guidelines, polypharmacy, and drug-drug


interactions in patients with multimorbidity, BMJ: British Medical
Journal (Online) 350.

[4] N. P. Tatonetti, P. Y. Patrick, R. Daneshjou, R. B. Altman, Datadriven


prediction of drug effects and interactions, Science translational
medicine 4 (125) (2012) 125ra31–125ra31.

[5] N. P. Tatonetti, G. H. Fernald, R. B. Altman, A novel signal detection


algorithm for identifying hidden drug-drug interactions in adverse

15
event reports, Journal of the American Medical Informatics Association
19 (1) (2011) 79–85.

[6] A. Rajkomar, J. Dean, I. S. Kohane, Machine learning in medicine., The New


England journal of medicine 380 14 (2019) 1347–1358.

[7] Y. Ba¸stanlar, M. Ozuysal, Introduction to machine learning, in: miR- ¨


Nomics: MicroRNA Biology and Computational Analysis, Springer, 2014,
pp. 105–128.

[8] G. Handelman, H. Kok, R. Chandra, A. Razavi, M. Lee, H. Asadi, ed octor:


machine learning and the future of medicine, Journal of internal
medicine 284 (6) (2018) 603–619.

[9] R. Miotto, F. Wang, S. Wang, X. Jiang, J. T. Dudley, Deep learning for


healthcare: review, opportunities and challenges, Briefings in
bioinformatics 19 (6) (2017) 1236–1246.

[10] C. Xiao, E. Choi, J. Sun, Opportunities and challenges in developing deep


learning models using electronic health records data: a systematic
review, Journal of the American Medical Informatics Association 25 (10)
(2018) 1419–1428.

[11] A. B. Raies, V. B. Bajic, In silico toxicology: computational methods for


the prediction of chemical toxicity, Wiley Interdisciplinary Reviews:
Computational Molecular Science 6 (2) (2016) 147–172.

[12] I. J. Onakpoya, C. J. Heneghan, J. K. Aronson, Worldwide withdrawal of


medicinal products because of adverse drug reactions: a systematic
review and analysis, Critical reviews in toxicology 46 (6) (2016) 477–
489.

[13] M. D. Segall, C. Barber, Addressing toxicity risk when designing and


selecting compounds in early drug discovery, Drug discovery today 19
(5) (2014) 688–693.

[14] K. Roy, S. Kar, R. N. Das, Understanding the basics of QSAR for


applications in pharmaceutical sciences and risk assessment, Academic
press, 2015.

16
[15] G. Patlewicz, J. M. Fitzpatrick, Current and future perspectives on the
development, evaluation, and application of in silico approaches for
predicting toxicity, Chemical research in toxicology 29 (4) (2016) 438–
451.

[16] Z. Y. Algamal, M. H. Lee, A. M. Al-Fakih, M. Aziz, High-dimensional qsar


prediction of anticancer potency of imidazo [4, 5-b] pyridine derivatives
using adjusted adaptive lasso, Journal of Chemometrics 29 (10) (2015)
547–556.

[17] J. M. Luco, F. H. Ferretti, Qsar based on multiple linear regression and


pls methods for the anti-hiv activity of a large group of hept derivatives,
Journal of chemical information and computer sciences 37 (2) (1997)
392–401.

[18] R. Tibshirani, Regression shrinkage and selection via the lasso, Journal
of the Royal Statistical Society: Series B (Methodological) 58 (1) (1996)
267–288.

[19] Z. Algamal, M. Lee, A new adaptive l1-norm for optimal descriptor


selection of high-dimensional qsar classification model for anti-hepatitis
c virus activity of thiourea derivatives, SAR and QSAR in Environmental
Research 28 (1) (2017) 75–90.

[20] J. Ma, R. P. Sheridan, A. Liaw, G. E. Dahl, V. Svetnik, Deep neural nets as a


method for quantitative structure–activity relationships, Journal of
chemical information and modeling 55 (2) (2015) 263–274.

[21] Y.-C. Lo, S. E. Rensi, W. Torng, R. B. Altman, Machine learning in


chemoinformatics and drug discovery, Drug discovery today.

[22] J. Shi, G. Zhao, Y. Wei, Computational qsar model combined molecular


descriptors and fingerprints to predict hdac1 inhibitors,
m´edecine/sciences 34 (2018) 52–58.

[23] E. L. Willighagen, J. W. Mayfield, J. Alvarsson, A. Berg, L. Carlsson, N.


Jeliazkova, S. Kuhn, T. Pluskal, M. Rojas-Chert´o, O. Spjuth, et al., The
chemistry development kit (cdk) v2. 0: atom typing, depiction,
molecular formulas, and substructure searching, Journal of
cheminformatics 9 (1) (2017) 33.

17
[24] T. Liu, Y. Lin, X. Wen, R. N. Jorissen, M. K. Gilson, Bindingdb: a
webaccessible database of experimentally determined protein–ligand
binding affinities, Nucleic acids research 35 (suppl 1) (2006) D198–
D201.

[25] A. Gaulton, L. J. Bellis, A. P. Bento, J. Chambers, M. Davies, A. Hersey, Y.


Light, S. McGlinchey, D. Michalovich, B. Al-Lazikani, et al., Chembl: a
large-scale bioactivity database for drug discovery, Nucleic acids
research 40 (D1) (2011) D1100–D1107.

[26] M. Nekoei, M. Mohammadhosseini, E. Pourbasheer, Qsar study of vegfr2


inhibitors by using genetic algorithm-multiple linear regressions
(gamlr) and genetic algorithm-support vector machine (ga-svm): a
comparative approach, Medicinal Chemistry Research 24 (7) (2015)
3037–3046.

[27] Z. Algamal, M. Qasim, H. Ali, A qsar classification model for


neuraminidase inhibitors of influenza a viruses (h1n1) based on
weighted penalized support vector machine, SAR and QSAR in
Environmental Research 28 (5) (2017) 415–426.

[28] H. W. Ng, S. W. Doughty, H. Luo, H. Ye, W. Ge, W. Tong, H. Hong,


Development and validation of decision forest model for estrogen
receptor binding prediction of chemicals using large data sets, Chemical
research in toxicology 28 (12) (2015) 2343–2351.

[29] K. Lee, M. Lee, D. Kim, Utilizing random forest qsar models with
optimized parameters for target identification and its application to
targetfishing server, BMC bioinformatics 18 (16) (2017) 567.

[30] I. Grenet, K. Merlo, J. P. Comet, R. Tertiaux, D. Rouqui´e, F. Dayan, Stacked


generalization with applicability domain outperforms simple qsar on in
vitro toxicological data, Journal of Chemical Information and Modeling.

[31] A. Lysenko, A. Sharma, K. A. Boroevich, T. Tsunoda, An integrative


machine learning approach for prediction of toxicity-related drug safety,
Life science alliance 1 (6) (2018) e201800098.

18
[32] K. M. Gayvert, N. S. Madhukar, O. Elemento, A data-driven approach to
predicting successes and failures of clinical trials, Cell chemical biology
23 (10) (2016) 1294–1301.

[33] V. Law, C. Knox, Y. Djoumbou, T. Jewison, A. C. Guo, Y. Liu, A. Maciejewski,


D. Arndt, M. Wilson, V. Neveu, et al., Drugbank 4.0: shedding new light
on drug metabolism, Nucleic acids research 42 (D1) (2013) D1091–
D1097.

[34] G. Consortium, et al., The genotype-tissue expression (gtex) pilot


analysis: multitissue gene regulation in humans, Science 348 (6235)
(2015) 648–660.

[35] K. J. Karczewski, B. Weisburd, B. Thomas, M. Solomonson, D. M. Ruderfer,


D. Kavanagh, T. Hamamsy, M. Lek, K. E. Samocha, B. B. Cummings, et al.,
The exac browser: displaying reference data information from over 60
000 exomes, Nucleic acids research 45 (D1) (2016) D840–D845.

[36] Y. Xu, Z. Dai, F. Chen, S. Gao, J. Pei, L. Lai, Deep learning for drug-induced
liver injury, Journal of chemical information and modeling 55 (10)
(2015) 2085–2093.

[37] D. Jimenez-Carretero, V. Abrishami, L. Fern´andez-de Manuel, I. Palacios,


A. Qu´ılez-Alvarez, A. D´ıez-S´anchez, M. A. del Pozo, M. C. Mon- ´ toya,
Tox (r) cnn: Deep learning-based nuclei profiling tool for drug toxicity
screening, PLoS computational biology 14 (11) (2018) e1006238.

[38] W. Torng, R. B. Altman, 3d deep convolutional neural networks for


amino acid environment similarity analysis, BMC bioinformatics 18 (1)
(2017) 302.

[39] Q. Hu, M. Feng, L. Lai, J. Pei, Prediction of drug-likeness using deep


autoencoder neural networks, Frontiers in genetics 9.

[40] A. Kadurin, S. Nikolenko, K. Khrabrov, A. Aliper, A. Zhavoronkov, drugan:


an advanced generative adversarial autoencoder model for de novo
generation of new molecules with desired molecular properties in silico,
Molecular pharmaceutics 14 (9) (2017) 3098–3104.

19
[41] H. Altae-Tran, B. Ramsundar, A. S. Pappu, V. Pande, Low data drug
discovery with one-shot learning, ACS central science 3 (4) (2017) 283–
293.

[42] J. E. Ridings, The thalidomide disaster, lessons from the past, in:
Teratogenicity Testing, Springer, 2013, pp. 575–586.

[43] C. A. Naranjo, U. Busto, E. M. Sellers, P. Sandor, I. Ruiz, E. Roberts, E.


Janecek, C. Domecq, D. Greenblatt, A method for estimating the
probability of adverse drug reactions, Clinical Pharmacology &
Therapeutics 30 (2) (1981) 239–245.

[44] J. Venulet, A. Ciucci, G. Berneker, Updating of a method for causality


assessment of adverse drug reactions., International journal of clinical
pharmacology, therapy, and toxicology 24 (10) (1986) 559–568.

[45] T. B. Agbabiaka, J. Savovi´c, E. Ernst, Methods for causality assessment


of adverse drug reactions, Drug safety 31 (1) (2008) 21–37.

[46] T. Sakaeda, A. Tamon, K. Kadoyama, Y. Okuno, Data mining of the public


version of the fda adverse event reporting system, International journal
of medical sciences 10 (7) (2013) 796.

[47] J. M. Banda, L. Evans, R. S. Vanguri, N. P. Tatonetti, P. B. Ryan, N. H. Shah,


A curated and standardized adverse drug event resource to accelerate
drug safety research, Scientific data 3 (2016) 160026.

[48] L. Hazell, S. A. Shakir, Under-reporting of adverse drug reactions, Drug


safety 29 (5) (2006) 385–396.

[49] D. S. Wishart, Y. D. Feunang, A. C. Guo, E. J. Lo, A. Marcu, J. R. Grant, T.


Sajed, D. Johnson, C. Li, Z. Sayeeda, et al., Drugbank 5.0: a major update
to the drugbank database for 2018, Nucleic acids research 46 (D1)
(2017) D1074–D1082.

[50] M. Kuhn, M. Campillos, I. Letunic, L. J. Jensen, P. Bork, A side effect


resource to capture phenotypic effects of drugs, Molecular systems
biology 6 (1) (2010) 343.

[51] M. Kuhn, I. Letunic, L. J. Jensen, P. Bork, The sider database of drugs and
side effects, Nucleic acids research 44 (D1) (2015) D1075–D1079.

20
[52] A. Gaulton, A. Hersey, M. Nowotka, A. P. Bento, J. Chambers, D. Mendez,
P. Mutowo, F. Atkinson, L. J. Bellis, E. Cibri´an-Uhalte, et al., The chembl
database in 2017, Nucleic acids research 45 (D1) (2016) D945–D954.

[53] J. Hastings, G. Owen, A. Dekker, M. Ennis, N. Kale, V. Muthukrishnan, S.


Turner, N. Swainston, P. Mendes, C. Steinbeck, Chebi in 2016: Improved
services and an expanding collection of metabolites, Nucleic acids
research 44 (D1) (2015) D1214–D1219.

[54] S. Kim, J. Chen, T. Cheng, A. Gindulyte, J. He, S. He, Q. Li, B. A. Shoemaker,


P. A. Thiessen, B. Yu, et al., Pubchem 2019 update: improved access to
chemical data, Nucleic acids research 47 (D1) (2018) D1102–D1109.

[55] G. Joshi-Tope, M. Gillespie, I. Vastrik, P. D’Eustachio, E. Schmidt, B. de


Bono, B. Jassal, G. Gopinath, G. Wu, L. Matthews, et al., Reactome: a
knowledgebase of biological pathways, Nucleic acids research 33 (suppl
1) (2005) D428–D432.

[56] M. Kanehisa, M. Furumichi, M. Tanabe, Y. Sato, K. Morishima, Kegg: new


perspectives on genomes, pathways, diseases and drugs, Nucleic acids
research 45 (D1) (2016) D353–D361.

[57] T. Lorberbaum, M. Nasir, M. J. Keiser, S. Vilar, G. Hripcsak, N. P. Tatonetti,


Systems pharmacology augments drug safety surveillance, Clinical
Pharmacology & Therapeutics 97 (2) (2015) 151–158.

[58] K. Raja, M. Patrick, J. T. Elder, L. C. Tsoi, Machine learning workflow to


enhance predictions of adverse drug reactions (adrs) through druggene
interactions: Application to drugs for cutaneous diseases, Scientific
reports 7 (1) (2017) 3690.

[59] K. Sornalakshmi, G. Vadivu, G. Sujatha, D. Hemavathi, A survey on using


social media data analytics for pharmacovigilance, Research Journal of
Pharmacy and Technology 10 (10) (2017) 3474–3478.

[60] A. Lusci, G. Pollastri, P. Baldi, Deep architectures and deep learning in


chemoinformatics: the prediction of aqueous solubility for drug-like
molecules, Journal of chemical information and modeling 53 (7) (2013)
1563–1575.

21
[61] M. Herrero-Zazo, M. Lille, D. J. Barlow, Application of machine learning
in knowledge discovery for pharmaceutical drug-drug interactions., in:
KDWeb, 2016.

[62] J. Zhao, Temporal weighting of clinical events in electronic health


records for pharmacovigilance, in: Bioinformatics and Biomedicine
(BIBM), 2015 IEEE International Conference on, IEEE, 2015, pp. 375–
381.

[63] J. Bekker, A. Hommersom, M. Lappenschaar, J. Davis, Measuring adverse


drug effects on multimorbity using tractable bayesian networks, arXiv
preprint arXiv:1612.03055.

[64] R. Moghaddass, C. Rudin, D. Madigan, The factorized self-controlled case


series method: An approach for estimating the effects of many drugs on
many outcomes, Journal of Machine Learning Research 17 (185) (2016)
1–24.

65] M. Morel, E. Bacry, S. Ga¨ıffas, A. Guilloux, F. Leroy, Convsccs:


convolutional self-controlled case series model for lagged adverse event
detection, arXiv preprint arXiv:1712.08243.

[66] Z. Kuang, Y. Bao, J. Thomson, M. Caldwell, P. Peissig, R. Stewart, R. Willett,


D. Page, A machine-learning-based drug repurposing approach using
baseline regularization, in: Computational Methods for Drug
Repurposing, Springer, 2019, pp. 255–267.

[67] Y. Xu, Y. Xu, S. Saria, A bayesian nonparametric approach for estimating


individualized treatment-response curves, in: Machine Learning for
Healthcare Conference, 2016, pp. 282–300.

[68] S. Haneuse, M. Daniels, A general framework for considering selection


bias in ehr-based studies: what data are observed and why?, eGEMs 4
(1).

[69] A. B. Abacha, M. F. M. Chowdhury, A. Karanasiou, Y. Mrabet, A. Lavelli, P.


Zweigenbaum, Text mining for pharmacovigilance: Using machine
learning for drug name recognition and drug–drug interaction
extraction and classification, Journal of biomedical informatics 58
(2015) 122–132.

22
[70] J. Mower, D. Subramanian, T. Cohen, Learning predictive models of drug
side-effect relationships from distributed representations of literature-
derived semantic predications, Journal of the American Medical
Informatics Association 25 (10) (2018) 1339–1350.

[71] H. H. Kim, K. Y. Rhew, A machine learning approach to classification of


case reports on adverse drug reactions using text mining of expert
opinions, in: Advances in Computer Science and Ubiquitous Computing,
Springer, 2017, pp. 1072–1077.

[72] T. Mikolov, K. Chen, G. Corrado, J. Dean, Efficient estimation of word


representations in vector space, arXiv preprint arXiv:1301.3781.

[73] I. Sutskever, O. Vinyals, Q. V. Le, Sequence to sequence learning with


neural networks, in: Advances in neural information processing
systems, 2014, pp. 3104–3112.

[74] M.-T. Luong, H. Pham, C. D. Manning, Effective approaches to attention-


based neural machine translation, arXiv preprint arXiv:1508.04025.

[75] D. Britz, A. Goldie, M.-T. Luong, Q. Le, Massive exploration of neural


machine translation architectures, arXiv preprint arXiv:1703.03906.

[76] A. Vaswani, N. Shazeer, N. Parmar, J. Uszkoreit, L. Jones, A. N. Gomez, L.


Kaiser, I. Polosukhin, Attention is all you need, in: Advances in Neural
Information Processing Systems, 2017, pp. 5998–6008.

[77] Y. Luo, W. K. Thompson, T. M. Herr, Z. Zeng, M. A. Berendsen, S. R.


Jonnalagadda, M. B. Carson, J. Starren, Natural language processing for
ehr-based pharmacovigilance: a structured review, Drug safety 40 (11)
(2017) 1075–1089.

[78] S. Dev, S. Zhang, J. Voyles, A. S. Rao, Automated classification of adverse


events in pharmacovigilance, in: Bioinformatics and Biomedicine
(BIBM), 2017 IEEE International Conference on, IEEE, 2017, pp. 1562–
1566.

[79] S. Zhang, S. Dev, J. Voyles, A. S. Rao, Attention-based multi-task learning


in pharmacovigilance, in: 2018 IEEE International Conference on
Bioinformatics and Biomedicine (BIBM), IEEE, 2018, pp. 2324–22328.

23
[80] F. Li, W. Liu, H. Yu, Extraction of information related to adverse drug
events from electronic health record notes: Design of an end-to-end
model based on deep learning, JMIR medical informatics 6 (4).

[81] X. Yang, J. Bian, Y. Gong, W. R. Hogan, Y. Wu, Madex: A system for


detecting medications, adverse drug events, and their relations from
clinical notes, Drug safety (2019) 1–11.

[82] A. Sarker, R. Ginn, A. Nikfarjam, K. OConnor, K. Smith, S. Jayaraman, T.


Upadhaya, G. Gonzalez, Utilizing social media data for
pharmacovigilance: a review, Journal of biomedical informatics 54
(2015) 202–212.

[83] A. C. Tricco, W. Zarin, E. Lillie, S. Jeblee, R. Warren, P. A. Khan, R. Robson,


G. Hirst, S. E. Straus, et al., Utility of social media and crowd-intelligence
data for pharmacovigilance: a scoping review, BMC medical informatics
and decision making 18 (1) (2018) 38.

84] S. Comfort, S. Perera, Z. Hudson, D. Dorrell, S. Meireis, M. Nagarajan, C.


Ramakrishnan, J. Fine, Sorting through the safety data haystack: Using
machine learning to identify individual case safety reports in
socialdigital media, Drug safety 41 (6) (2018) 579–590.

[85] R. A. Calix, R. Gupta, M. Gupta, K. Jiang, Deep gramulator: Improving


precision in the classification of personal health-experience tweets with
deep learning, in: Bioinformatics and Biomedicine (BIBM), 2017 IEEE
International Conference on, IEEE, 2017, pp. 1154–1159.

[86] A. Cocos, A. G. Fiks, A. J. Masino, Deep learning for pharmacovigilance:


recurrent neural network architectures for labeling adverse drug
reactions in twitter posts, Journal of the American Medical Informatics
Association 24 (4) (2017) 813–821.

[87] J. Liu, G. Wang, Pharmacovigilance from social media: An improved


random subspace method for identifying adverse drug events,
International Journal of Medical Informatics 117 (2018) 33–43.

[88] A. Nikfarjam, A. Sarker, K. Oconnor, R. Ginn, G. Gonzalez,


Pharmacovigilance from social media: mining adverse drug reaction
mentions using sequence labeling with word embedding cluster

24
features, Journal of the American Medical Informatics Association 22 (3)
(2015) 671–681.

[89] Z. Wu, W. Li, G. Liu, Y. Tang, Network-based methods for prediction of


drug-target interactions, Frontiers in pharmacology 9.

[90] D. Dana, S. Gadhiya, L. St Surin, D. Li, F. Naaz, Q. Ali, L. Paka, M. Yamin, M.


Narayan, I. Goldberg, et al., Deep learning in drug discovery and
medicine; scratching the surface, Molecules 23 (9) (2018) 2384.

[91] F. Hammann, V. Sch¨oning, J. Drewe, Prediction of clinically relevant


drug-induced liver injury from structure using machine learning,
Journal of Applied Toxicology.

[92] L. Zhang, H. Zhang, H. Ai, H. Hu, S. Li, J. Zhao, H. Liu, Applications of


machine learning methods in drug toxicity prediction, Current topics in
medicinal chemistry 18 (12) (2018) 987–997.

[93] M. Benhenda, Chemgan challenge for drug discovery: can ai reproduce


natural chemical diversity?, arXiv preprint arXiv:1708.08227.

[94] K. Preuer, P. Renz, T. Unterthiner, S. Hochreiter, G. Klambauer, Fr´echet


chemblnet distance: A metric for generative models for molecules, arXiv
preprint arXiv:1803.09518.

[95] Y. LeCun, B. E. Boser, J. S. Denker, D. Henderson, R. E. Howard, W. E.


Hubbard, L. D. Jackel, Handwritten digit recognition with a back-
propagation network, in: NIPS, 1989.

25
TABLE 1: Open-source Databases with molecular, or pharmacological
information
Name Description References Resource

DrugBank a bioinformatics and [33,49] xi


chemoinformatics resource
about drug data and drug
target
ChEMBL a manually curated database [25,52] vi
of bioactive molecules with
drug-like properties
SIDER a database on marketed [50,51] xvi
medicines and their recorded
adverse drug reactions
ChEBI a freely available dictionary [53] xx
of molecular entities focused
on “small” chemical
compounds
PubChem an open chemistry database [54] xxi
at the National Institutes of
Health (NIH)
Reactome a curated and peer-reviewed [55] xxii
pathway database
KEGG a database resource for [56] xxiii
understanding high-level
functions and utilities of the
biological system, the
organism and the ecosystem,
from molecular-level
information

Figure Legend

Figure 1: Artificial intelligence and machine learning present a opportunity


for improving drug safety. These algorithms enable a data-driven approach

26
to toxicity and safety assessments that can identify patterns that otherwise
would be overlooked. Traditional machine learning, including methods like
logistic regression, random forests, and support vector machines can
produce interpretable models with relatively low complexity. These methods
are desirable when the goal is to understand how the predictors affect the
incidence or risk of an adverse event. A new class of methods, called deep
neural networks – and often referred to as “artificial intelligence” – allows for
more complex models to be built at the cost of requiring significantly more
data. The benefit of using these algorithms is that they can automatically
identify non-linear patterns in the data without requiring much manual
intervention. Common examples that have been used in drug safety research
include convolutional and recurrent neural networks. In both cases, these
models have been used to in pre-clinical drug toxicity study, to model patient
diversity, and to facilitate lead selection and trial design, and in post-marking
surveillance to conduct comparative effectiveness research, identify drug-
drug interactions, and to aid in clinical decision making. AI-assisted drug
safety and toxicity science remains a nascent and growing field that requires
further research to evaluate its potential clinical impact.

27
GLOSSARY

Attention mechanism: a process that allows to look at elements in a


sequence as a whole and learn a distribution that weights their
contextual importance.

AC50: Concentration for half-maximal activity, as derived from the Hill


equation. It is a common potency measured employed in toxicity testing.

Area under the Receiver Operating Curve (AUC): Area between the curve
and the x-axis. Heuristic used to evaluate the performance of
classification models with AUC =1 indicating perfect classification.

Bayesian methods: Bayesian and Frequentist are two different approaches


to defining probabilities. The Bayesian approach is to see as a
representation of uncertainty, while frequentists see probabilities as a
long-term frequency of an event.

Bayesian non-parametrics (BNP): a class of methods where the complexity


of a model is defined by the data.

Convolutional neural networks (ConvNet or CNN): were pioneered by


LeCun et al. in 1990 in their first application for handwritten digit
recognition. They consist of training filters that performs convolutional
products with the input data and learn more and more high level
features. ConvNets have to learn comparatively less weights than fully
connected neural networks, and are particularly efficient for computer
vision applications.

Data dimensionality: Refers to the number of attributes present in a dataset

Deep learning (DL): Deep learning (DL) is a sub-field of machine


learning where the algorithms learn abstractions of the input
features that they use to make the predictions. These algorithms
are characterized by a higher capacity than classic machine
learning techniques (i.e., have higher degrees of freedom). It is in
essence the trade-off of DL: what we gain in capacity and
automated feature engineering, we lose it in a higher dimension
space of parameters that is more complex and time-consuming to
explore.

28
Diffusion state distance (DSD): Distance metric based on properties of
graph diffusion designed to capture distinctions between annotations
in protein-protein interactions

Embedding of semantic predications (ESP): a method to generate


semantic vector representations of biomedical terms inspired by the
skipgram-with-negative-sampling (SGNS). SGNS is an embedding
method that uses neural networks to associate terms to their context
in a corpus.

Genetic algorithm (GA): GA is a stochastic variable selection method that


solves optimization problems by applying Darwinian hypotheses of
evolution.

Gradient boosting classifier: An approach used for classification and


regression that builds a predictive model using a combination of
individually weaker prediction models.

k-fold cross validation: A resampling procedure in which the data is split


into k groups in order to estimate and assess model performance.

k-nearest neighbor (k-NN): Algorithm used for classification in which the


data are separated into several classes to help predict the classification
of a new data point

Latent factor analysis (LFA): A statistical method used to describe


variability in observed and correlated variables in term of unobserved
variables called latent factors.

LD50 (Lethal dose 50%): Amount of an administered substance that kills


50% of a test sample.

Linear discriminant analysis (LDA): Statistical, machine learning


technique which seeks to find a linear combinate of features that
separates two or more classes.

Machine learning (ML): a field of artificial intelligence in which


algorithms are trained to perform tasks and make predictions by learning
directly from the data, without being explicit programmed. ML methods
can broadly be classified into two classes based on how the data learn to

29
make predictions: supervised and unsupervised learning. In supervised
learning, an algorithm is used to learn the mapping between input
variables and an output, such as a label. The goal is for the algorithm to
learn to predict a correct output when a new input is provided. In
unsupervised learning, there are no assigned labels to the input training
data. Here, the machine’s goal is to learn representations of the input data
that can be used for tasks such as predicting future inputs, and decision
making, without an output.

Naïve bayes: set of supervised learning algorithm based on Bayes


theorem with the assumption of conditional independence between
feature pairs

Named Entity Recognition (NER): a method that identified tokens in


unstructured text and map them to concepts or categories in
terminologies.

Natural Language Processing (NLP): Subfield of computer science that


aims at manipulating and making sense of natural language data.

Random forest: An ensemble learning approach in classification and


regression which constructs decision trees during training and produces
the class (classification) or mean predication (regression) of individual
trees.

Regression: Statistical approach which seeks to find relationships be-


tween dependent variables and one or more independent variables.
Multivariate regression estimates a single regression model with more
than one outcome variable.

Self-controlled case series (SCCS): A method in epidemiological study
design where the subjects are their own control.

Undirected Graph Recursive Neural Networks (UG-RNN): In
chemoinformatics, a model where, for a molecule of N atoms, a multi-
layer perceptron (MLP) recursively crawls through N different
representations of this molecule. The vectors generated as a result are
then averaged to compute a prediction for the molecule.

30
Word embeddings: In Natural Language Processing (NLP), one of the main
challenges is finding a good representation for the vocabulary the
corpus covers. While some methods simply encode tokens in binary
vectors with a sparse representation, word embeddings learn a
representation that takes into account the token's context

31

You might also like