Artificial Intelligence in Drug Discovery: Applications and Techniques

Download as pdf or txt
Download as pdf or txt
You are on page 1of 65

Artificial Intelligence in Drug Discovery:

Applications and Techniques

Jianyuan Deng,∗,† Zhibo Yang,‡ Iwao Ojima,¶ Dimitris Samaras,‡ and Fusheng
arXiv:2106.05386v4 [cs.LG] 2 Nov 2021

Wang†,‡

†Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, USA
‡Department of Computer Science, Stony Brook University, Stony Brook, NY, USA
¶Department of Chemistry, Stony Brook University, Stony Brook, NY, USA

E-mail: [email protected]

Abstract

Artificial intelligence (AI) has been transforming the practice of drug discovery

in the past decade. Various AI techniques have been used in many drug discovery

applications, such as virtual screening and drug design. In this survey, we first give an

overview on drug discovery and discuss related applications, which can be reduced to

two major tasks, i.e., molecular property prediction and molecule generation. We then

present common data resources, molecule representations and benchmark platforms.

As a major part of the survey, AI techniques are dissected into model architectures

and learning paradigms. To reflect the technical development of AI in drug discovery

over the years, the surveyed works are organized chronologically. We expect that this

survey provides a comprehensive review on AI in drug discovery. We also provide a

GitHub repository with a collection of papers (and codes, if applicable) as a learning

resource, which is regularly updated.

1
Introduction

Drug discovery is well known as an expensive, time-consuming process, with low success
rates. On average, developing a new drug costs 2.6 billion US dollars 1 and can take more
than 10 years. Moreover, the success rate of launching a drug to market from Phase I
clinical trial is daunting, less than 10%. 2 In the past decade, the practice of drug discovery
has been undergoing radical transformations driven by the rapid development in artificial
intelligence (AI). 3–7 Popular applications of AI in drug discovery include virtual screening, 8
de novo drug design, 9 retrosynthesis and reaction prediction, 10 and de novo protein design, 11
among others, which can be reduced to two categories, i.e., predictive and generative tasks.
To power these AI applications, a wide range of AI techniques are involved, with model
architectures evolving from traditional machine learning models to deep neural networks,
such as convolutional neural networks, recurrent neural networks, graph neural networks and
transformers, etc. Learning paradigms also shift from supervised learning to self-supervised
learning and reinforcement learning.
In this survey, we focus on the applications and techniques of AI-driven discovery on
small-molecule drugs. Biologics (e.g. antibodies, vaccines) are not covered. We first provide
an overview of key applications in drug discovery and point out a collection of previously
published perspectives, reviews, and surveys. We then introduce common data resources and
representations of small molecules. We also discuss existing benchmark platforms for both
molecular property prediction and molecule generation. With knowledge on data and repre-
sentations, the related techniques, including model architectures and learning paradigms, will
be elaborated. Finally, we discuss existing challenges and highlight some future directions.
By assembling a Github repository 1 with the surveyed papers (and codes, if applicable), we
expect this survey not only provides a comprehensive overview of AI in drug discovery but
also serves as a learning resource for researchers interested in the interdisciplinary field.
1
https://github.com/dengjianyuan/Survey_AI_Drug_Discovery

2
Drug Discovery Overview

In this section, we first go over the definitions of key concepts in drug discovery, mainly
on the screening and design of small molecules. Note that AI-powered drug repositioning,
which repurposes existing drugs or drug combinations for new indications, is not included.
Besides, target identification, exploiting -omics data for its druggability, is also out of the
scope and thus not discussed. Rather, we refer the readers to previous publications on drug
repositioning 12,13 and -omics data for target identification. 14
Drug discovery 15 is a project motivated by the situation when there are no drugs for a
disease or when existing drugs have limited efficacy and/or severe toxicity. At the earliest
stage, an underlying hypothesis needs to be developed that activation or inhibition of a
target (e.g., an enzyme, a receptor, an ion channel, etc) results in therapeutic effects for the
disease, which involves target identification and target validation. For the selected target,
intensive assays will be performed to find the hits and subsequently the leads (i.e., drug
candidates), which involves hit discovery, hit-to-lead phase and lead optimization. The drug
candidates then enter preclinical studies and clinical trials. If successful, the drug candidate
can be launched into market as a medical product to treat the disease.
To accelerate the small-molecule drug discovery, high-throughput screening (HTS) 16,17
has been proposed to increase the discovery efficiency since the 1980s, which is a hit-finding
approach underpinned by development in automation and the availability of large chemical
libraries. A prominent outcome of HTS is the large-scale structure-activity relationship
(SAR) datasets, which contribute to the chemical databases such as PubChem 18 and ZINC. 19
Various computational techniques have been developed to search the chemical libraries for
potentially active molecules to be tested in subsequent in vitro and in vivo assays, 20 which is
also known as virtual screening (VS). In other words, VS is to identify active molecules using
computational approaches, based on knowledge about the target (structure-based VS ) or
known active ligands (ligand-based VS ) to increase the odds of identifying active molecules. 21
For the concept of active molecules, as mentioned above, activation or inhibition of a target

3
is the underlying hypothesis to treat a disease, which corresponds to two major classes of
drugs with regard to the mechanism of action (MoA), i.e., agonists and antagonists. 22 An
agonist is a molecule which activates the target to exert a biologic response as its endogenous
ligand. On the contrary, an antagonist is a molecule which binds to the target to block the
response. Based on more specific effects and mechanisms, agonists can further be classified
as partial agonists, inverse agonists, biased agonists. Antagonists include competitive and
non-competitive antagonists. To quantify the activity, various assays have been developed
to measure affinity (or potency) and efficacy. Affinity is the fraction or extent to which a
molecule binds to a target at a given concentration whereas potency is the necessary amount
of a molecule to produce an effect of a given magnitude, inversely proportional to the affinity.
Efficacy, on the other hand, describes the effect size, such as inhibition of an enzyme to 60%.
Common activity measures are summarized in Table 1.
Table 1: Common Measures of Molecule Activity

Measures Definition
Kd Equilibrium dissociation constant
Km Michaelis constant
Ki Inhibition constant
IC50 Half maximal inhibitory concentration
EC50 Half maximal effective concentration

Nevertheless, sufficient activity is not the only criterion for an ideal drug candidate, which
only makes it a ligand. Binding specificity is another concern. 23 Most often, a molecule can
bind to multiple targets and unexpected side effects may arise due to binding promiscuity.
Thus, high selectivity is another desired feature. In fact, drug candidates should also sat-
isfy a combination of criteria, 24 with optimal physicochemical (water solubility, acid-base
dissociation constant, lipophilicity, permeability), pharmacokinetic (absorption, distribution,
metabolism, excretion), and pharmacodynamic (activity, selectivity) properties. Other prop-
erties considered during compound synthesis include the Synthetic Accessibility Score (SAS)
and the Quantitative Estimation of Drug-likeness (QED). SAS is a heuristic score of how hard
(10) or easy (1) it is to synthesize a given molecule based on a combination of the molecular

4
fragments’ contributions. QED is an estimate (0-1) on how likely a molecule is a viable drug
candidate. At its core, drug discovery is a multi-objective optimization problem. 25 Usually,
for each property of interest, a predictive model is built to map the molecular structure to
the property value with either classification or regression, which is broadly referred to as
quantitative structure-activity relationship (QSAR) modeling. 26 A more intriguing prospect
from QSAR is that these models can be exploited inversely to reveal the structural features
underlying the optimal properties to guide drug design from scratch, also known as de novo
drug design.
Rather than merely screening existing chemical libraries, 27 drug design takes a step
further to explore the vast chemical space, i.e., the space encompassing all possible small
molecules 28 which has an estimated size around 1030 − 1060 . 29 In drug design, there is a
design-make-test-analysis (DMTA) cycle, which consists of iterative organic synthesis and
property assays. 3 To efficiently navigate the chemical space, quantitative drug design has
been proposed since late 1970s. 30 Essentially, drug design epitomizes in two questions: 31 1)
“Can molecular properties be deduced from molecular structures?” and 2) “Which structural
features are relevant for certain molecular properties?” The former one also underlies the
core assumption of VS and the latter is what QSAR tries to answer. In this sense, drug
design can be viewed as an extension to VS, and involves both molecular property prediction
and molecule generation, which are the major tasks in current AI-driven drug discovery.

Summary of Existing Reviews

Next, we briefly discuss existing reviews on AI-driven discovery by dividing them into three
categories: 1) “General drug discovery review”, 2) “Drug discovery in the AI-era”, and 3)
“Rethinking AI-driven drug discovery”.

5
General Drug Discovery Reviews

Many existing papers have covered the general aspects of drug discovery 3,15 and related
concepts, such as chemical space, 28 VS and HTS, 21,32,33 optimal properties for drug can-
didates, 24 QSAR, 26,34 target prediction, 35 and computer-aided drug design. 27 Besides, one
prominent challenge in drug discovery is that molecular properties can be highly sensitive
to minor structural changes. This is also known as the activity cliffs (ACs), where pairs
of structurally similar molecules exhibit significantly different activities. 36–38 We strongly
recommend the readers (especially those new to drug discovery) refer to these reviews for a
better understanding on drug discovery and recognition of potential pitfalls.

Drug Discovery in the AI Era

AI has been widely applied in drug discovery. Since the early 2000s, machine learning
models, such as random forest (RF), have been exploited for VS and QSAR. 39,40 In 2012,
AlexNet 41 marked the advent of the deep learning era. 42 Shortly after in the 2012 Merck
Kaggle competition, deep neural networks (DNN) outperformed the standard RF model in
predicting molecular activities. 39 More recently, the success of AI techniques in computer
vision and natural language processing has shed more light into drug discovery 4,6,43,44 and
led to the burgeoning field of deep learning in chemistry. 5 In 2019, potent inhibitors of dis-
coidin domain receptor 1 (DDR1) were discovered in 21 days by researchers from Insilico
Medicine. 45 In 2020, a novel antibiotic candidate against antibiotic-resistant bacteria, halicin,
was identified by researchers from MIT. 46 Note that AI can be applied at different stages in
drug discovery, from target identification and validation to drug response determination. 6
Lead identification, the focus of this survey, involves two fundamental tasks, i.e., molecular
property prediction and molecule generation. Molecular property prediction, at the core of
VS, is to predict the property value of a molecule given its structure or learned represen-
tation, 47 which can be served for various purposes, such as drug-target interaction (DTI)
prediction, 35 toxicity prediction 48 and drug-induced liver injury (DILI) prediction, 49 among

6
others. Molecule generation, underlying drug design, involves two levels of tasks: 1) realistic
molecule generation, i.e., generating molecules within constraints imposed by the chemical
rules, and 2) goal-directed molecule generation, i.e., generating chemically valid molecules
with desired properties. 50,51

Rethinking AI-driven Drug Discovery

Despite the promise of AI in drug discovery, pitfalls still exist, which have been widely
discussed. 8,9,31,52–55 As opined by Bender et al, 53,54 “a method cannot save an unsuitable
representation which cannot remedy irrelevant data for an ill thought-through question”. To
circumvent potential hypes and unrealistic expectations thereof, there is indeed a necessity to
take into consideration the hypotheses, the data, the representations, the models, the learning
paradigms and moreover, these components as a whole, for any AI-driven application in drug
discovery.

Structure of the Survey

Given the necessity of a clear understanding on both drug discovery applications and AI
techniques, this survey starts from general aspects in drug discovery and then moves on to AI-
driven drug discovery, covering data resources, molecule representations, model architectures,
and learning paradigms. The organization of this survey is depicted in Fig 1.
Notably, current literature on the AI techniques is often fragmented. More often than not,
the rationale behind the choice of a technique is simply because it has not been previously
investigated. 51 To gain more insights into the strengths and weaknesses of these AI tech-
niques, we focus on the model architectures and learning paradigms. We also try to present
the surveyed works chronologically so as to reflect the technical development over the years.
Finally, we highlight existing challenges and future directions (see Section “Discussion”).

7
Application Task Representation Architecture Learning Paradigm

Images Self-Supervised
CNN
Drug Design Molecular Learning
Property RNN
Prediction Few-Shot Learning
SMILES GNN
Quantitative Strings Metric Learning
Structure VAE
Activity
Relationship Meta Learning
Molecular GAN
Graphs
Active Learning
Flow
Molecule
Virtual Generation
Screening Fixed Reinforcement
Transformer
Fingerprints Learning

Figure 1: Applications and Techniques of AI in Drug Discovery. The applications of AI


in small-molecule drug discovery include virtual screening, quantitative structure-activity
relationship and drug design, which can be reduced to two major tasks: molecular prop-
erty prediction and molecule generation. Small molecules can be represented by fixed fin-
gerprints, molecular graphs, simplified molecular input entry system (SMILES) strings, and
images. Various model architectures have been applied on each representation format, includ-
ing convolutional neural networks (CNN), recurrent neural networks (RNN), graph neural
networks (GNN), variational autoencoders (VAE), generative adversarial networks (GAN),
normalizing flow models and transformers. Still, challenges exist for the low-data molecu-
lar property prediction and goal-directed molecule generation. To tackle these challenges,
different learning paradigms have been proposed, such as self-supervised learning for the
pretraining-finetuning practice and reinforcement learning for navigating the chemical space
search. Other paradigms surveyed here also include few-shot learning, metric learning, meta
learning and active learning.

Data, Representation and Benchmark Platforms

In this section, we first discuss the publicly available data resources. Then, we discuss how
small molecules can be represented in machine-readable formats. Lastly, we summarize cur-
rent benchmark platforms for both molecular property prediction and molecule generation.

Public Data Resources

With the improvements in HTS and related assays, data on molecular activity and related
properties are ever increasing, which contribute to various public data resources. These
resources typically provide information on molecular structures, molecular properties and
target information, 56 as discussed below.

8
PubChem 57 was launched by the National Institutes of Health in 2004. With a collection
of chemical information from 750 data sources, PubChem is the largest chemical database. As
of August 2020, PubChem contains 111 million unique chemical structures with 271 million
activity data points from 1.2 million biological assays experiments. PubChem provides direct
download as well as web interfaces for online queries. Notably, PubChem is non-curated 56
and the bioactivity datasets from PubChem can be highly imbalanced. 58 Researchers may
curate the data on their own. For example, Chithrananda et al 59 recently released a curated
dataset of 77 million SMILES strings from PubChem. ChEMBL, 60 maintained by the Euro-
pean Molecular Biology Laboratory, is another large-scale chemical database. For example,
in ChEMBL22 (version 22), there are more than 1.6 million distinct chemical structures with
over 14 million activity values. Moreover, ChEMBL is manually curated in a comprehen-
sive manner. 56 ChEMBL provides downloads in a variety of formats (e.g., Oracle, MySQL
or PostgreSQL database) and also allows web application program interface (API) for data
retrieval in XML or JSON format. 61 Notably, based on ChEMBL, Mayr et al 62 extracted
a large-scale benchmark dataset for target prediction. ZINC, 19 developed by the Irwin and
Shoichet Laboratories in UCSF, contains a suite of molecules, annotated ligands and targets
as well as the purchasability for over 120 million “drug-like” compounds. ZINC supports
direct download from the website and also provides an API for retrieving data. Notably,
some subsets of the ZINC database are more commonly used, such as the ZINC-250k 63 and
the ZINC Clean Leads collections. 64
In addition to the aforementioned large-scale databases, there are also other data reposito-
ries, 65 such as PDBbind, BindingDB, DUD, DUD-E, MUV, STITCH, GLL&GDD, NRLiST
BDB, KEGG, among others. Besides databases mainly derived from preclinical studies, there
are also public data resources for the marketed drugs and their effects in human subjects,
such as the datasets for adverse drug reactions (ADR) (e.g., DrugBank, SIDER, OFFSIDES
and TWO-SIDES) and the datasets for DILI ( e.g., DILIrank). 66

9
Small Molecule Representations

Molecules are often depicted as Kekulé diagrams with bonds and atoms (Fig 2A). Over the
years, machine-readable representations have been developed to enable rapid computation,
querying and storage of the molecules. 67 Molecules can be represented by the fixed molecu-
lar descriptors, which are further categorized by its dimensionality. 56 Specifically, there are
0D descriptors for molecules, such as molecular weight (MW), atom number, and atom-
type count. 0D descriptors can be directly derived from the empirical formula and barely
provide information on how atoms are connected. For example, the empirical formula of
alanine, C3 H7 N O2 , can also represent lactamide. 67 To highlight different functional groups,
descriptors incorporating more structural information have been proposed, such as finger-
prints (Fig 2B). Fingerprints are binary vectors with each dimension in the vector indicating
the presence or absence of a particular substructure. Among them, there are 1D descriptors
to represent the substituent atoms, chemical bonds, structural fragments, and functional
groups. There are also 2D descriptors to represent the atom connectivity and molecular
topology, such as 1) Keyed fingerprints - molecular access system (MACCS) keys, 2) Path-
based fingerprints - DayLight fingerprints and 3) Circular fingerprints - extended connectivity
fingerprints (ECFPs) based on the Morgan algorithm. 68 Furthermore, 3D descriptors have
also been developed to encode 3D-structural information like the steric properties, surface
area, volume and binding site properties, among others.
Molecular descriptors have greatly boosted the application of computational methods,
including machine learning models, in drug discovery. 69,70 Nevertheless, these descriptors
are fixed and not learnable towards improving model performance. With the advent of the
AI era, various deep learning models have paved the way for end-to-end (E2E) predictions,
where molecules can be embedded into a continuous latent space without hand-crafted rules.
Among them, two major representation formats are molecular graphs and the simplified
molecular input entry system (SMILES) strings. 67

10
Node Indices

Node Features

Node Indices
Node Indices
ur es
eat
geF
Ed
A. Kekulé Diagram C. Molecular Graph Node Feature Matrix Adjacency Tensor

[0 0 0 1 … 0 0 0 0 1]

Tokenization

One-Hot Encoding
B. Fingerprints D. SMILES String
C ( = O ) c 1

Figure 2: Illustration of Small Molecule Representations.

Molecular Graphs

The idea of graph representation is intuitive, where atoms are typically mapped to nodes
and bonds to edges. Formally, a graph is defined as G = (V, E), a set of of nodes (atoms)
V and a set of edges (bonds) E, where (vi , vj ) ∈ E indicates a bond between atoms vi and
vj . 67 The attributes of atoms are represented by a node feature matrix X and each node
v can be represented by an initial vector xv and a hidden vector hv ∈ RD . Similarly, the
attributes of bonds can also be represented by an edge feature matrix. Note that both node
and edge feature matrices do not directly encode connections. Instead, an adjacency matrix
A keeps track of the pairwise connection status. The element of A, aij , if equals to 1, means
that there is a bond connecting node vi and vj ; otherwise, if aij equals to 0, these two nodes
are not connected by a bond. Usually, the edge feature matrix and the adjacency matrix are
combined to form an adjacency tensor (Fig 2C). Common node and edge features 71,72 are
summarized in Table 2.
One advantage of the graph representations is that they carry more structural infor-
mation. Besides, molecular graphs as well as the subgraphs can be directly mapped to a
chemical (sub-)structure and thus are highly interpretable. 73 One drawback of the graph

11
Table 2: Common Node and Edge Features in Molecular Graphs

Type Feature Notes


Node Atom type Element type
Node Formal charge Assigned charges
Node Implicit Hs Number of bonded hydrogens
Node Chirality R or S configuration
Node Hybridization Orbital hybridization: spx , spx dy
Node Aromaticity Aromatic atom or not
Edge Bond type Single, double, triple or aromatic
Edge Conjugated Conjugated or not
Edge Stereoisomers cis (Z) or trans (E)

representation, however, is that these matrices require a large amount of disk space for stor-
age and significant memory during computation, which may slow down the efficiency during
molecule generation. 67

SMILES Strings

To accommodate the storage and computation efficiency, molecules are also commonly repre-
sented by the SMILES strings. 74 In SMILES, an atom is represented by the atomic symbols;
for two-character symbols, the second letter will be represented in lower case. Elements in
the organic subset, namely B, C, N, O, P, S, F, Cl, Br and I, can be written without brackets
whereas for those not included, brackets should be applied with the attached hydrogens and
formal charges written inside, such as [Fe2+]. The lower-case letters represent the atoms in
aromatic rings; for instance, C is used for the normal carbon and c is used for the aromatic
carbon. For bonds, there are single, double, triple and aromatic bonds, represented by the
symbols -, =, # and :, respectively, where single bonds and aromatic bonds are usually
omitted. For the branches in a molecule, they are denoted by enclosures in parentheses. To
represent the cyclic structure, a single or aromatic bond is first broken down in the ring and
then the bonds are numbered in any order with the ring-opening bonds by a digit following
the atomic symbol at each ring. Notably one molecule may correspond to multiple SMILES
strings. 67 To avoid conflicts, canonicalization methods 75 have been introduced to ensure only

12
one unique SMILES string is designated for the same molecule.
Usually the SMILES string are converted into one-hot vectors before fed into the machine
learning models (Fig 2D). 76 Comparing to the graph representation, SMILES string is less
computationally expensive. However, since SMILES strings do not directly encode the atomic
connection, there can be a loss of the structural information. 77 Besides, due to the internal
syntax of the SMILES (e.g., ring opening and closure, atom valency), using this linear
notations for molecule generation is prone to generate invalid molecules. 78,79

Other Representations

Molecules can also be represented by more sophisticated 3D-atomic coordinates, commonly


seen in structure-based VS or QSAR studies. 80–83 Molecular topology, such as bond lengths,
bond angles and torsional angles, can also be incorporated. 84 Some works have already
exploited the 3D-representation to generate molecules. 85,86 In addition to the raw 3D co-
ordinates, well-established 3D properties, which capture the molecular conformation, can
also be readily utilized for prediction tasks. 87 Furthermore, with the advances of computer
vision, images of molecular structures (Fig 2A) emerge as another modality to represent
molecules. 88–92

Benchmark Platforms

To evaluate the performance of molecular property prediction and molecule generation, there
are several benchmark platforms, which are discussed below.
As a major benchmark dataset platform for molecular property prediction, MoleculeNet
was released by Wu et al in 2018, 93 which includes a set of datasets along with an open-source
DeepChem package. 94 The benchmark datasets cover four categories: 1) Quantum mechan-
ics (QM7, QM7b, QM8, QM9), 2) Physical chemistry (ESOL, FreeSolv, Lipophilicity), 3)
Biophysics (PCBA, MUV, HIV, PDDBind, BACE), and 4) Physiology (BBBP, Tox21, Tox-
Cast, SIDER, ClinTox), involving single task or multi tasks. Notably, for molecular property

13
prediction, datasets can be highly imbalanced. Thus, when choosing evaluation metrics (Ta-
ble 3), positive rates should be considered. For instance, AUPRC is favored over AUROC
in case of a low positive rate (e.g. less than 2%). 93 With regard to the datset splitting,
in addition to the common random split, MoleculeNet also provides other splitting ways,
namely, scaffold split, stratified split and time split for different datasets. In other words,
for each dataset, the recommended split way varies. For example, for the BACE dataset,
since it is for a single target, the scaffold splitting is more suitable, whereas for the PDBind
dataset, since the data collection is over a long period, time splitting is recommended to
better reflect the actual drug discovery effort over the years.
One limitation of MoleculeNet, however, is that it does not provide explicit training,
validation and test folds for the datasets. 95 To improve reproducibility, the ChemBench
package from MolMapNet 96 was released recently. MolMapNet also expands the Molecu-
leNet by adding pharmacokinetics-related datasets, such as PubChem CYP inhibition and
liver microsomal clearance data. In addition to the benchmark datasets, Chemprop, 71 for
benchmarking the learned molecular representations, was proposed in 2019, which system-
atically compared the fixed molecular descriptors (e.g. ECFPs) and the learned molecular
representations for molecular property prediction. In Chemprop, models were benchmarked
extensively on 19 public and 16 proprietary industrial datasets. As a side note, Chemprop
is related to the discovery of halicin. 46
As for benchmarking the molecule generation models, Olivecrona et al 97 developed REIN-
VENT in 2017, which is a sequence-based generative model utilizing SMILES strings. REIN-
VENT can be used to execute a range of tasks, such as generating analogues to a query struc-
ture and generating ligands for a given target. In 2020, Blaschke et al 98 proposed the updated
version, REINVENT 2.0, as a production-ready tool for drug design. For benchmarking
molecule generation utilizing molecular graphs, Mercado et al 72 proposed GraphINVENT
in 2020. To standardize the assessment for molecule generation, an evaluation framework
GuacaMol 99 was proposed in 2019, which set a suite of tasks for distribution learning and

14
goal-directed design. More specifically, the generative model is examined on whether it can
reproduce the property distribution of training sets (usually for VS purpose 100 ), and find
the optimal molecule (for multi-objective optimization). A more recent evaluation platform
MOSES was released by Polykovskiy et al in 2020, 64 which compiles a list of metrics (Table 3
2
) for detecting common issues in generative models, such as overfitting and mode collapse.

Table 3: Commonly Used Evaluation Metrics

Application Task Metric Purpose


Virtual screening Molecular property prediction Recall@k Retrieval
Virtual screening Molecular property prediction Precision@k Retrieval
Virtual screening Molecular property prediction AP@k Retrieval
QSAR Molecular property prediction Accuracy Classification
QSAR Molecular property prediction Recall Classification
QSAR Molecular property prediction Precision Classification
QSAR Molecular property prediction AUROC Classification
QSAR Molecular property prediction AUPRC Classification
QSAR Molecular property prediction MAE Regression
QSAR Molecular property prediction RMSE Regression
Drug design Molecule generation Validity Distribution learning
Drug design Molecule generation Unique@k Distribution learning
Drug design Molecule generation Novelty Distribution learning
Drug design Molecule generation Diversity Distribution learning
Drug design Molecule generation FCD Distribution learning
Drug design Molecule generation KL divergence Distribution learning
Drug design Molecule generation Scaffold similarity Goal-directed design
Drug design Molecule generation Rediscovery Goal-directed design

For instance, validity measures how well a model explicitly captures the chemical rules,
such as valency; uniqueness and diversity examine whether the generative model collapses to
producing only a limited set of molecules; novelty indicates whether the model is overfitted
to just memorize the training examples. Furthermore, Fréchet ChemNet Distance (FCD) is a
measure of how close the distributions of the generated set are to the distribution of molecules
in the training set. A low FCD value corresponds to similar molecule distributions. Kullback-
2
QSAR: quantitative structure-activity relationship; Recall@k: recall among top k molecules; Preci-
sion@k: precision among top k molecules; AP@k: average precision among top k molecules; AUROC:
area under the receiver-operating characteristic curve; AUPRC: area under the precision-recall curve; MAE:
mean absolute error; RMSE: rooted mean square error; Unique@k: uniqueness of the first k valid (generated)
molecules; FCD: Fréchet ChemNet Distance; KL divergence: Kullback-Leibler divergence.

15
Leibler (KL) divergence measures the difference between two probability distributions. When
the KL divergence value is small, the generated molecules can well approximate the targeted
property in the training set. For goal-directed design, it relies on a formalism where molecules
are scored individually based on each pre-defined criterion, such as containing a specific sub-
structure, having certain physicochemical properties or exhibiting similarity or dissimilarity
to certain molecules. Consequently, similarity and rediscovery are usually used for evaluation
purpose. Specifically, rediscovery assesses if the generative model is able to rediscover a
given molecule and similarity evaluates whether the model can generate molecules similar or
dissimilar to a given molecule.

Model Architectures

Prior to the “deep learning” era, traditional machine learning models were widely used in
VS. 40 Pertinent tasks include predictions of drug likeliness, 101,102 physicochemical proper-
ties, 103,104 pharmacokinetic parameters 105–108 and pharmacodynamic properties. 109,110 There
are score-based classification models, support vector machines (SVM) 111 and K nearest neigh-
bors (KNN) and probability-based classification models, random forest (RF), 112 naive bayes
(NB), and logistic regression (LR). Despite the success of traditional machine learning mod-
els, deep neural networks (DNNs) have outperformed them in a variety of tasks. 62,113

Convolutional Neural Networks

Convolutional neural networks (CNNs) are mainly used in computer vision to process pixels
of data in images. 114 In CNNs, there are convolution layers and pooling (i.e. subsampling)
layers (Fig 3). On top of these convolution layers and pooling layers, a vector representation
is learned by concatenating the feature maps for a final prediction. CNNs share parameters
across the filters, which largely reduces the number of parameters to be learned, thereby
decreasing memory consumption and increasing computation speed.

16
Pooling Concatenation
y
Convolution
Pooled Learned
Input Image Feature Maps Feature Maps Vector

Figure 3: Illustration of Convolutional Neural Networks.

In drug discovery, CNNs can be applied to elucidate the bioactivity profiles based on
microscopy images. 115,116 Moreover, CNNs are also used for molecular property predic-
tion. 117,118 In 2015, Duvenaud et al 118 applied CNNs on circular fingerprints, a refinement
of the ECFPs, 119 to create a differentiable fingerprint, which is among the first efforts us-
ing data-driven representation learning for molecular property prediction, instead of fixed
chemical descriptors. This work has greatly motivated learning molecular representations.
In addition to fingerprints, CNNs can also effectively extract features directly from the
images of molecular structure. For instance, Chemception 120 is trained on the 2D-structural
images to predict free energy of solvation and inhibition of HIV replication. Later, Fernández
et al 88 developed Toxic Colors, a framework for toxicity classification with the images as in-
put. Cortes-Ciriano et al 90 further extended existing CNNs architectures (e.g. AlexNet, 41
DenseNet-201, 121 ResNet152 122 and VGG-19 123 ) to Kekulé structure images for molecular
property prediction, also known as KekuleScope. The experimental results of KekuleScope
showed that CNNs on images as input can achieve comparable performance to RF and DNNs
on ECFPs. Meyer et al 89 also predicted the MeSH-therapeutic-use classes based on com-
pound images, which outperformed previous predictions based on transcriptomic data. More
recently, Rifaioglu et al 91 proposed a large-scale DTI prediction system, DEEPScreen. In-
deed, molecular property prediction with images as input are closely related to the progress
in computer vision, which also prompts automatic extraction of chemical structures from
literature and patents. 92,124 The chemical structure recognition model can be further inte-

17
grated with models in natural language processing, such as DECIMER 125 and DECIMER
1.0, 92 which are able to translate the bitmap images of a molecule into a SMILES string, as
an image captioning task. 126

Recurrent Neural Networks

Recurrent neural networks (RNNs) are mainly used for processing sequential data. 114 RNNs
allow the connection among neurons in the same hidden layer to form a directed cycle
(Fig 4A), thereby enabling the use of sequential input, such as language modeling 127 and
music generation. 128 If unfolded in time steps, RNNs can be seen as a very deep feed-forward
networks where all layers sharing the same weights. However, the long-term dependency of
RNNs makes it difficult to learn the parameters due to the gradient explosion or vanish-
ing problem. 114 As a result, long short-term memory (LSTM) 129 and gated recurrent unit
(GRU), 130 two variants of the vanilla RNN, have been developed to augment the network
with a memory module. Different from CNNs’ operation on images, RNNs mainly take the
SMILES strings as input for molecular property prediction and molecule generation. As
discussed in Section “Small Molecule Representation”, the characters in a SMILES string
are firstly converted into one-hot vectors (Fig 2) and then sequentially fed into RNNs, with
a hidden vector to be updated at each step. For molecular property prediction, RNNs gen-
erate a final output after all steps are taken. For example, SMILES2Vec 131 uses RNNs to
learn features from SMILES and predicts a wide range of chemical properties. Mayr et al 62
proposed SmilesLSTM to perform DTI prediction, which outperformed traditional machine
learning models.
RNNs can also be applied for molecule generation, similar to language models for text gen-
eration. 97,100,132 More specifically, RNNs generate output at each step in an auto-regressive
manner (Fig 4B), where the output is dependent on the input from previous steps. Based on
the input from the current step and prior steps, RNNs output a probability distribution over
all possible tokens, from which a token is sampled as the output of the current step and will

18
y x' End
y
Token

unfold unfold
h h0 h1 h2 hT h h0 h1 h2 hT

Learned
x x1 x2 xT x
Vector
Input Sequence Sequentially Input Input Sequence Start
Token

A. Recurrent Neural Networks in Prediction Mode B. Recurrent Neural Networks in Generation Mode

Figure 4: Illustration of Recurrent Neural Networks.

be used to predict the next token. However, due to the syntax of the “SMILES language”
such as the ring opening-closure and the matched brackets, regular RNNs, including LSTM
and GRU, cannot capture the algorithmic patterns of the sequence well owing to their in-
ability to count. 79 As a result, the generated SMILES strings very often violate the chemical
rules and become invalid. Thus, a memory-augmented version, Stack-RNN, 79,133 was devel-
oped to alleviate the validity issue for SMILES-based molecule generation. Another solution
for this problem is to adopt bidirectional RNNs, such as the bidirectional LSTM. 134,135
In addition to the SMILES strings, RNNs can also be applied on molecular graphs for
generation purpose. 136–139 For example, You et al 136 proposed GraphRNN to generate molec-
ular graphs in an autoregressive manner, decomposing it as a process into generating a se-
quence of node and edge formations conditioned on the graph structure generated so far.
Nonetheless, generating molecular graphs with RNNs requires a full trajectory of the graph
generation, which tends to forget the states of initial generation steps quickly. Later You et
al proposed GCPN and 140 designed the graph generation procedure as a Markov Decision
Process (MDP), which only needs the intermediate state to generate the graph. Notably,
RNNs can also be components of more complicated generative models, such as variational
autoencoders 78,141 and generative adversarial networks. 142,143

19
Graph Neural Networks

CNNs and RNNs are usually applied on data represented in the Euclidean space. In recent
years, graph neural networks (GNNs) are gaining popularity to model data represented in
graphs with a set of nodes and edges. 144 GNNs can handle node-level (e.g., node classifica-
tion), edge-level (e.g., link prediction) and graph-level (e.g., graph regression) tasks, with
neighborhood aggregation, pooling and readout operations. Small molecules, when repre-
sented as molecular graphs (Fig 2C), are naturally appealing to the application of GNNs for
both molecular property prediction and molecule generation tasks (see Section “Molecular
Graphs)”.
Two major types of GNNs are convolutional GNNs (ConvGNNs) and recurrent GNNs. 144
In the recurrent GNNs, node representation is learned via some recurrent neural architec-
tures, such as the graph gated neural network (GGNN). 145 On the other hand, ConvGNNs
generalize the convolution operation from grid data to graph data and can stack multiple
graph convolutional layers to extract high-level node representations. ConvGNNs play a
central role in building up many other complex GNNs, which can be further categorized into
two subtypes: 1) Spectral-based: ChebNet, 146 graph convolutional network (GraphConv) 147
and 2) Spatial-based: message passing neural networks (MPNN), 148 GraphSAGE, 149 graph
attention network (GAT), 150 graph isomorphism network (GIN). 151
In drug discovery, GNNs are often exploited for molecular property prediction (Fig 5A).
For instance, Kearnes et al 152 developed Weave to perform graph convolutions on molecular
graphs for representation learning, where the graph convolutions, nonetheless, did not out-
perform the fingerprint-based models back in 2016. Later, Gilmer et al 148 proposed MPNN
as a unified framework for quantum chemical properties prediction. MPNN has two phases
in the forward pass, namely, message passing and readout. During message passing, for each
atom, feature vectors from its neighbors are propagated into a message vector wherein the
hidden vector for the atom is updated by the message vector. A readout function is used to
aggregate the feature vectors into a graph feature vector, which is then passed to a fully con-

20
Message Passing Readout
y

Learned
Vector
Node Feature Vector Neighborhood Aggregation: k hops in L iterations

A. Graph Neural Networks in Prediction Mode

Adjacency Tensor
Node Feature Matrix

Initialization Sampling
G0

Empty Graph Initial Atom


Append Connect Terminate

B. Graph Neural Networks in Generation Mode

Figure 5: Illustrations of Graph Neural Networks.

nected layer for downstream predictions. Yang et al 71 then expanded MPNN into directed
MPNN (D-MPNN), which uses messages associated with directed edges (bonds) instead of
nodes (atoms) used in MPNN, thereby preventing repeated message passing from the same
node. Notably, Yang et al 71 also introduced a practice to concatenate the 200 global molec-
ular features calculated by RDKit 153 with the learned features by D-MPNN for downstream
predictions, also adopted in later works. 63 Xiong et al 77 integrated graph attention mecha-
nism into GNNs and developed Attentive FP, which is able capture topologically adjacent
atoms’ interactions for improved molecular property prediction. More recently, Withnall
et al 154 also made augmentations to the MPNN and proposed attention MPNN (AMPNN)
and edge memory neural network (EMNN) for physicochemical property prediction. So far,
GNNs have been widely applied for molecular property prediction. More examples include
as SchNet, 155 PotentialNet, 156 and DimeNet, 157 among others. 62,158–165 Moreover, subgraphs
can directly map to molecular substructures, which also improves interpretability. 77,166–168
Partly encouraged by the superior performance of GNNs for molecular property predic-
tion, GNNs are also exploited for molecule generation (Fig 5B). As mentioned above, RNNs

21
can be used to generate molecular graphs, which, nevertheless, needs to store a full trajec-
tory for the graph generation process and tends to forget initial states. 140 In 2018, Li et
al 138 developed a conditional graph generator, MolMP, which does not involve atom-level
recurrent units. MolMP models the graph generation as a MDP problem, where the action
to grow graph only depends on its current state. There are three actions in total - append,
connect and terminate, the sampling process of which is parameterized by a neural network.
Experimental results show that MolMP outperforms SMILES-based molecule generation in
a variety of evaluation metrics, especially the validity. GNNs-based molecule generation can
be used in common drug design applications such as designing molecules with certain scaf-
folds, presumably due to more straightforward mapping to chemical substructure with the
graph representation. Furthermore, since molecule generation is usually driven by certain
desired properties, reinforcement learning (see Section “Learning Paradigms”), therefore,
is often integrated with GNNs for goal-directed drug design. Examples include GCPN, 140
MolDQN, 169 DeepGraphMolGen, 170 and MNCE-RL. 171 For more practical issues on GNNs
for molecule generation, such as generation schemes (single-shot vs iterative) and computa-
tion, we refer the readers to the guide by Mercado et al. 51

Variational Autoencoders

Variational autoencoders (VAEs), a class of powerful probablistic generative models, were


first introduced by Kingma et al 172 in 2013. VAEs, consisting of an encoder E and a decoder
D. The encoder maps high-dimensional data into a low-dimensional, continuous latent space
(Fig 6). Compared to common autoencoders, the latent space is regularized to be organized,
ideally, through the KL divergence. In addition to reconstruction, VAEs approximate a
probability distribution, which can be sampled for generation purpose. Thus, given input
x, the parameters of VAEs are optimized by minimizing the reconstruction loss and the KL
divergence: 173
||x − D(E(x))||2 + KL N (µx , σx ), N (0, 1) ,

(1)

22
which is equivalent to maximize the evidence lower bound (ELBO). In Equation 1, N (0, 1)
denotes the unit normal distribution; µx and σx are learnable parameters, representing mean
and variance of a Gaussian distribution.

x Latent Space x'


Gaussian Assumption

Encoder Decoder
p(z|x) p(x|z)

Sampling

Molecules with Targeted Properties

Figure 6: Illustration of Variational Autoencoders.

VAEs can be applied on SMILES strings for molecule generation. For example, Gómez-
Bombarelli et al 78 developed a VAE model for automatic molecule design, where a pair of
deep neural networks (i.e. an encoder and a decoder) is trained as an autoencoder to convert
the input SMILES strings into a continuous vector representation. To train the autoencoder,
a reconstruction loss is adopted in attempt to reproduce the original SMILES string. How-
ever, the ultimate goal is not to merely reconstruct the input. Rather, the autoencoder
aims to learn a compact representation for the molecules. Thus, a constraint is applied in
the autoencoder by jointly training a physical property regression model to organize the
VAE’s latent space subjected to the property value, which can be used to sample molecules
towards the desired property value. Partly due to the syntax of SMILES, the latent space
learned by the autoencoder can be sparse and contain large “dead areas”, which correspond
to invalid molecules. Therefore, VAEs with a focus on the syntax for valid molecule genera-
tion are proposed later, such as GrammarVAE 174 and syntax-directed VAE. 175 Other related
works also include semi-supervised VAE (SSVAE) for continuous output, 176 conditional VAE
(CVAE), 177 constrained graph VAE (CGVAE), 178 NeVAE, 179 GTM VAE 141 and CogMol. 180
A variant of the VAE is the adversarial autoencoder (AAE), 181 which replaces the KL diver-
gence with an adversarial objective. More specifically, the Gaussian distribution assumption

23
as a prior on the latent representations for KL-divergence computation is replaced by other
priors, i.e., an additional discriminator is added to force the encoder generates latent repre-
sentations in a specific distribution (e.g. a uniform distribution). AAEs can also be used for
molecule generation, 182–184 which can improve reconstruction and the validity of generated
molecules.
VAEs can also be applied on the graph representations for generation purpose. In
2018, Simonovsky et al 185 proposed a VAE framework (GraphVAE) to generate molecular
graphs.Their main idea is to output a probabilistic fully-connected graph and use a standard
graph matching algorithm to align it to the ground truth. Jin et al 186 developed the junction
tree VAE (JT-VAE). In JT-VAE, a molecular graph is first mapped into a junction tree via
a tree decomposition algorithm and the junction tree then undergoes the VAE’s encoding-
decoding process. The learned latent space of the junction tree can be used to search for
substructures, which then assemble into molecules with specific properties. A prominent
merit of JT-VAE is that the validity of all generated molecules can be guaranteed. Ma et
al 187 also proposed a regularization framework for VAEs (Regularized VAE) that regular-
ize the output distribution of the decoder, thereby improving the validity. Later, Kajino
et al 188 developed molecular hypergraph grammar VAE (MHG-VAE), where a molecular
graph is described as a hypergraph and the grammar VAE 174 is trained by inputting the
grammar for sequence production of the hypergraph. In 2019, Kwon et al 189 developed a
non-autoregressive graph VAE and incorporated three additional learning objectives into the
model, namely, approximate graph matching, reinforcement learning, and auxiliary property
prediction, which is able to generate valid and diverse molecular graphs with various con-
straints. Moreover, graph-based VAEs can also embrace a strategy for drug design with the
ability to retain a particular scaffold (i.e., substructure), such as the ScaffoldVAE. 190
One drawback of VAEs, nonetheless, is that the set of substructures by partitioning
molecules can be quite large. Consequently, the iterative prediction of which substructure to
add can be inaccurate, especially for infrequent substructures. To address this challenge, Fu

24
et al 191 proposed a novel strategy, CORE, by combining scaffolding tree generation and ad-
versarial training. Besides, the computational cost increasing with the number of nodes in a
graph is another major challenge here, limiting the application to larger molecules. In 2020,
Kwon et al 192 proposed a compressed graph representation to alleviate computational com-
plexity while maintaining the validity and diversity of generated molecules. More recently,
Jin et al 193 developed hierarchical graph VAE (HierVAE) which can employ larger and more
flexible graph motifs as building blocks for molecules. More specifically, the encoder pro-
duces a multi-resolution representation for each molecule in a fine-to-coarse fashion, from
atoms to connected motifs while the autoregressive coarse-to-fine decoder adds one motif at
a time. Notably, HierVAE can even be used to generate polymers.

Generative Adversarial Networks

Generative Adversarial Networks (GANs), developed by Goodfellow et al 194 in 2014, have


made remarkable achievements in generating realistic synthetic samples. GANs consist of a
generative model G, and a discriminative model D (Fig 7). The generator aims to gener-
ate new data points from a random distribution whereas the discriminator aims to classify
whether the generated samples are from the training data distribution or from the generator.
GANs can be trained by the min-max loss, which alternatively optimizes the generator and
the discriminator using a min-max objective:

min max L(G, D) = Ex∼px [log(D(x))] + Ez∼pz [log(1 − D(G(z)))], (2)


G D

where px and pz denote the distribution of the real data x and the noise prior z.
GANs can be applied to SMILES strings for molecule generation. In 2017, Guimaraes et
al 142 developed objective-reinforced GANs (ORGAN), built upon SeqGAN, 195 to generate
molecules in SMILES strings while also optimizing several domain-specific metrics. The gen-
erator is based on LSTM, which is modeled as a stochastic policy in a reinforcement learning

25
Noise Generator
z G(x)
Generation
Latent Space
Discriminator y Real or not
D(x)

Real Molecule

Figure 7: Illustration of Generative Adversarial Networks.

setting (more details in Section “Learning Paradigms”), whereas the Wasserstein loss is
used train the discriminator (a CNN model). Experimental results showed that the gener-
ated molecules exhibit drug-like structures as well as improvement in the evaluation metrics.
Shortly after, an objective-reinforced GANs for inverse-design chemistry (ORGANIC) 143 was
developed based upon ORGAN. ORGANIC can generate molecules with biased distribution
towards certain attributes for both drug discovery and material design. In 2018, Putin et
al 196 presented a reinforced adversarial neural computer (RANC) framework, which also
combines GANs and RL. The generator of the RANC framework is a differentiable neural
computer (DNC) with an explicit memory bank, instead of the LSTM model in ORGANIC
and ORGAN. This is because the generation of discrete data using RNNs, particularly,
LSTM with maximum likelihood estimation, can suffer from the so-called “exposure bias”,
i.e., missing salient features of the data. RANC outperforms ORGANIC, as measured by
several metrics: number of unique structures, passing medicinal chemistry filters (MCFs),
Muegge criteria and high QED scores. RANC is able to generate molecules that match the
distributions of the key chemical features/descriptors (e.g., MW, logP) and lengths of the
SMILES strings from the training set.
GANs can also be applied on molecular graphs for molecule generation. In 2018, De Cao
et al 197 developed MolGAN, an implicit, likelihood-free generative model for small molecular
graph generation, which circumvents the expensive graph matching procedures. 185 Moreover,

26
they adapted the GANs to enable direct operation on molecular graphs. RL is also integrated
to encourage the generation of molecules towards desired properties. Experimental results on
the QM9 dataset showed that MolGAN is able to generate nearly 100% valid molecules, which
outperforms ORGAN in validity. One drawback of MolGAN is its susceptibility to mode
collapse, i.e., repeated samples being generated multiple times, leading to low uniqueness. 198

Normalizing Flow Models

In addition to the RNNs, VAEs and GANs, another major class of generative models is
the normalizing flow models. 199 Representative works include the Non-linear Independent
Component Estimation model (NICE), 200 the Real-valued Non-Volume Preserving model
(RealNVP), 201 and the Glow model, among others. In NICE, Dinh et al 200 introduced
tractable calculation for reversible transformations, which are also known as the affine cou-
pling layers underlying the flow models. The basic idea of flow models is to learn an invertible
mapping between complex distributions and simple prior distributions (Fig 8). By exploiting
exact and tractable likelihood estimation for training, flow models enable efficient one-shot
inference and 100% reconstruction of the training data. 51

Forward Inverse
Coupling zX Coupling

Node Feature Matrix X Node Feature Matrix X

Forward Inverse
Coupling zA Coupling

Adjacency Tensor A
Adjacency Tensor A

Figure 8: Illustrations of Flow Models.

In drug discovery, flow models have been applied to generate molecules, mainly on molec-
ular graphs. In 2019, Madhawa et al 202 developed GraphNVP, the first flow model for molec-
ular graph generation. In GraphNVP, the graph generation is decomposed into two steps,

27
i.e., generation of an adjacency tensor and generation of node attributes, which yields the
exact likelihood maximization on the graph with two reversible flows. GraphNVP is able to
generate valid molecules with minimal duplicates. The learned latent space can be further
exploited to generate molecules with desired properties. Honda et al 203 also developed an
invertible flow model for molecular graph generation based on residual flows, also known as
graph residual flow (GRF), which enables more flexible and complex non-linear mappings
than the traditional coupling flows. Experimental results showed that GRF can achieve
comparable performance with GraphNVP, while having much less parameters to learn. No-
tably, GraphNVP 202 and GRF 203 generate molecular graphs in a single-shot manner, 51 which
may lead to low validity, nevertheless. Consequently, a sequential iterative graph generation
manner is proposed for flow models. For example, GraphAF, 204 an autoregressive flow-based
model, adopts an iterative sampling process to leverage chemical domain knowledge, such as
valency checking in each step. With the integration of chemical rules, GraphAF is able to
generate molecules of 100% validity. Moreover, its training process is significantly faster than
GCPN. 140 GraphAF can be further finetuned with RL, which achieves better performance
on molecular property optimization compared to JT-VAE 186 and GCPN. 140 MoFlow, later
developed by Zang et al, 205 applies a validity correction to the generated graph, which not
only enables efficient molecular graph generation in a single-shot manner, but also guarantees
the chemical validity. The continuous latent space learned via encoding the molecular graphs
can be further used to generate novel and optimized molecules during the decoding process
towards desired properties. More recently, Luo et al 206 developed GraphDF, which, on the
contrary, aims to learn a discrete latent representation with the flow models and capture
the original discrete distribution of the discrete graph structures without adding real-valued
noise. For molecule generation, GraphDF sequentially samples the discrete latent variables
and maps them to new nodes and edges via invertible transforms. The discrete transforms
can circumvent the cost of computation while also achieving state-of-the-art performance in
random molecule generation, property optimization and constrained optimization tasks.

28
For the flow-based generative models, the most prominent feature is that they are able
to exactly reconstruct all the input data without duplicates due to the precise likelihood
maximization, which can be an important complement for molecule generation. In particular,
when the molecular property is highly sensitive to minor structural changes, i.e., activity
cliffs, 36,37 a replacement of a specific atom (node) might be needed. In other words, flow
models can offer more precise modifications on existing molecular structures.

Transformers

RNNs have been widely applied to handle sequential input. However, RNNs can suffer from
the gradients explosion or the vanishing problem. 114 In 2017, a seminal work “Attention is
all you need” proposed a novel transformer architecture, built with the self-attention mecha-
nism. 207 Transformers have now become the de facto standard in powerful language models,
such as GPT, 208 BERT, 209 GPT-2, 210 RoBERTa, 211 and GPT-3, 212 and even in advanced
computer vision models, such as DETR 213 and Vision Transformer. 214 Unlike RNNs, trans-
formers forfeit recurrent connections. By adopting positional embedding, transformers are
even better at dealing with long sequences. 207

CLS C C = O Contextual Property Prediction

Output Embedding

Input Output
Transformer Encoder

Positional Embedding Mask

Input Embedding
RDKit Motif

CLS C C Mask O Input Output

Tokenization Masking
Graph-Level Motif Prediction

A. Masked Language Modeling on SMILES Strings B. Context and Motif Prediction on Molecular Graphs

Figure 9: Illustrations of Self-Supervised Learning with Transformers.

Not unexpectedly, transformers are being actively applied in drug discovery. Notably,

29
transformers enable effective self-supervised pretraining, such as masked language model-
ing (Fig 9A). In 2019, Wang et al 215 developed SMILES-BERT, which consists of several
transformer encoder layers, to improve molecular property prediction. SMILES-BERT is
first pretrained on a large-scale corpus of SMILES strings via a SMILES recovery task and
then fine-tuned on the downstream prediction tasks. Later, Honda et al 216 proposed to
learn molecular representations through pretraining a sequence-to-sequence language model,
which is termed as the SMILES Transformer. Chithrananda et al 59 also developed Chem-
BERTa, built upon the RoBERTa model 211 for molecular property prediction. More recently,
Fabian et al 95 applied the architecture of BERT 209 to learn molecular representations, also
referred to as MolBERT. When pretrained with masked language modeling and other tasks,
MolBERT achieves improved performance for molecular property prediction compared to the
fixed fingerprints. Moreover, transformers can also be applied on molecular graphs, especially
considering that the transformer encoder can be viewed as a GAT variant. 150 For example,
Rong et al 63 developed a novel framework, GROVER, to learn graph representations with
the message passing transformer. By designing self-supervised contextual property predic-
tion and graph-level motif prediction tasks (Fig 9B), GROVER is pretrained on 10 million
unlabeled molecules and achieves state-of-the-art performance on 11 benchmark datasets.
In addition to molecular property prediction, transformers can also be exploited for
molecule generation, such as MoleculeChef, 217 which can generate the reactants for a given
product, similar to machine translation. More recently, transformers are also exploited for
protein-specific molecule generation, 218 where the input is the amino acid sequence of the
target protein and the output are ligands in the SMILES representation.

Learning Paradigms

Drug discovery, despite the light shed by AI, still faces major challenges. For molecular prop-
erty prediction, labeled data points are at the core of machine learning models. Nonetheless,

30
in real-world settings, generating labeled data points in wet lab can be very expensive. Con-
sequently, the datasets for model training are usually limited in size, exhibit high sparsity,
and can be heavily biased and noisy, which is also termed as the low-data drug discovery
problem. 158,165 For molecule generation, although existing generative models, such as VAEs,
can be used to generate molecules towards desired properties, the mechanism by mapping
from the points in the latent space to real molecules which are most proximal can limit the
exploration of the chemical space, leading to low novelty and diversity. 78 To address these
challenges, various learning paradigms have been proposed. In this survey, we mainly focus
on self-supervised learning and reinforcement learning to address molecular property predic-
tion and molecule generation, respectively. Other learning paradigms are also discussed.

Self-Supervised Learning

The performance of deep neural networks, especially supervised learning, hinges on a large
labeled dataset. Nevertheless, supervised learning is meeting its bottleneck due to its heavy
reliance on expensive manually-labeled data. 219 In real-world problems such as molecular
property prediction, the labeled data is often limited, sparse and biased, which leads to low
generalizalibility of models. Self-supervised learning is promising paradigm and has achieved
state-of-the-art performance in learning with limited labels, as adopted in the aforementioned
language models, for instance, BERT. 209 Notably, self-supervised learning should be distin-
guished from unsupervised learning. Unsupervised learning focuses on detecting patterns
in data without labels, such as clustering, whereas self-supervised learning aims to recover
the data. More specifically, it can be classified into two main types, i.e., generative and
contrastive self-supervised learning.
For the generative self-supervised learning, a canonical task is the masked language mod-
eling, as proposed in BERT, 209 where the model is trained to predict the masked tokens,
thereby recovering the original input. Model parameterization is usually implemented by
optimizing the cross-entropy loss between the output and the masked tokens in the in-

31
put. Representative works of self-supervised learning in drug discovery are discussed in Sec-
tion “Transformers” (e.g., MolBERT 95 and GROVER 63 ). Notably, self-supervised pretrain-
ing can avoid the negative transfer caused by supervised pretraining - transfer of knowledge
from pretraining harms model generalization, as shown by Hu et al. 163 Besides, contrastive
learning is another type of self-supervised learning. More specifically, contrastive learning
aims to learn latent representations through contrasting data pairs (positive vs negative),
where the positive and negative examples are constructed by augmenting the unlabeled
samples in a self-supervised manner. Recently, contrastive learning has been employed to
address the low-data drug discovery problem. For instance, Wang et al 220 proposed molecular
contrastive learning of representations (MolCLR) on molecular graphs for molecular prop-
erty prediction. Three molecular graph augmentation ways are used, i.e., atom masking,
bond deletion, and subgraph removal. Through a contrastive loss, MolCLR learns molec-
ular representations by contrasting positive vs negative molecules, where molecular graph
pairs augmented from the same molecule are treated as the positive and the others denoted
as negative. Experimental results show that MolCLR can effectively transfer the learned
representations to downstream tasks and achieve state-of-the-art performance in molecular
property prediction.
In addition to self-supervised learning, other learning paradigms have also been exploited
to address the low-data drug discovery challenge. For example, meta learning 221 aims to
learn a learner to be adapted to new tasks. In a study by Nguyen et al, 165 the meta-learning
initializations outperform multi-task pretraining baselines on 16 out of 20 in-distribution
tasks and all out-of-distribution tasks. A member of the meta-learning family is few-shot
learning, 222 the core idea of which is to generalize with a few examples. For instance, Altae
Tran et al 158 proposed a one-shot learning framework for activity classification, which lowers
the amount of data required for predictions. Intuitively, through learning a distance metric,
molecules can be embedded into the latent space in a more organized way. Thus, when
new molecules come, their embeddings can be compared to the exiting labeled molecules for

32
more accurate prediction. Closely to this idea, another paradigm is metric learning, 223,224
which mainly deals with data with mixed distribution due to the activity cliffs. 38 Metric
learning has been widely applied in computer vision, 224,225 especially in situations where
the similarity or distance must be computed for clustering or nearest neighbor classification
purpose. At its core, a distance metric (e.g., cosine distance) is to be learned, based on
which the learned latent representations of the input data can be separated according to
their labels. In a recent work by Na et al, 226 a generalized deep metric learning (GeDML)
framework is proposed, which alleviates the structure-property mismatch problem through
better separating molecules in the latent space. The representations learned via metric
learning are also conducive for goal-directed molecule generation, i.e., search in the chemical
space. For example, Koge et al 227 proposed a molecular embedding framework by combining
VAEs and metric learning. The idea is to make the molecules’ embedding in the latent space
consistent with their properties, thereby enabling efficient search during molecule generation.

Reinforcement Learning

With improved performance on molecular property prediction, another challenge still poses
for molecule generation, i.e., how to design molecules with the desired properties? As men-
tioned in Section “Model Architectures”, VAEs and flow models can be used to generate
molecules with preferred properties by sampling from a learned latent space. However, the
latent space can be highly dimensional and the objective functions defined in the latent space
is usually non-convex, making it difficult to optimize the properties of generated molecules. 169
Consequently, reinforcement learning (RL) is often used as the alternative to navigate the
chemical space, which mainly deals with how an agent should take actions in a certain state
so as to maximize a reward or return. 228 RL algorithms can be classified into 1) value-based
(e.g., Q-learning), 2) policy-based (e.g., policy gradient) and 3) hybrid (e.g., actor-critic). 229
In drug discovery, the DMTA cycle (see Section “Drug Discovery Overview”) under-
lying drug design, 3 i.e., goal-directed molecule generation, can be potentially automated

33
by RL through connecting the generative model (i.e., the agent for molecule generation)
and the predictive model (i.e., assigning rewards based on the predicted property values).
For example, Zhou et al 169 adopted value-based, double Q-learning (DQN) 230 to optimize
the generated molecules. Other related works mainly adopt the policy-gradient algorithm,
REINFORCE, 231 as an estimator of the gradient, such as ORGAN, 142 REINVENT, 97 and
ORGANIC. 143 Another policy-gradient algorithm, proximal policy optimization (PPO), 232
is also gaining popularity recently, which is improved from the trust region policy optimi-
sation (TRPO). 233 TRPO employs a trust region so that optimizations are restricted to a
region where the approximation of the true cost function holds, thereby preventing policies
updated too wildly and lowering the chance of a catastrophically “bad” update. 229 However,
TRPO requires the calculation of second-order gradients, being computationally expensive.
PPO, on the contrary, only requires first-order gradients and can retain the performance of
TRPO, exhibiting low sample complexity. Studies adopting PPO for de novo drug design
include the work by Neil et al, 132 GCPN, 140 DeepGraphMolGen 170 and MNCE-RL. 171
Nevertheless, policy-gradient algorithms usually exhibit high variance since the gradi-
ent estimation can be noisy. 229 To reduce the variance, an improved class of algorithms is
the hybrid actor-critic method, which combines policy-gradient methods with learned value
functions. For example, off-policy deterministic policy gradient (DPG) extends the stan-
dard policy gradients for stochastic policies to deterministic policies, which only integrates
over the state space instead of both state and action spaces, thus requiring fewer samples in
problems with large action spaces. Later, deep deterministic policy gradient (DDPG) utilizes
neural networks on high-dimensional space is introduced, as adopted in MolGAN. 197 Another
technique for variance minimization to accelerate convergence is to subtract the estimated
reward from the true reward, which separates the policy training from value estimation, also
known as the advantage actor-critic (A2C) algorithm adopted by Neil et al. 132
With RL training, chemical libraries shifted towards desired properties are expected to
be generated. However, drug design is a multi-objective optimization problem. 9,234 In order

34
to prioritize molecules based on the pre-defined goals, non-dominated sorting or pair-based
comparisons are usually exploited to find solutions with the Pareto optimality. 235–237 Another
issue with RL is the trade-off between exploration and exploitation. As illustrated by Zhou
et al, 169 this trade-off is a dilemma underlying by uncertainty. Due to the lack of a complete
knowledge of the rewards for all the states, if constantly choosing the best action known
to produce the highest reward (exploitation), the model will never learn anything about
the rewards of the other states; on the other hand, if always choosing a random action
(exploration), the model will not receive sufficient reward. A potential solution for the
exploration-exploitation trade-off is active learning, which is a paradigm where the model can
query an expert or any other information sources in an active manner during learning. 31,238

Discussions

There has been a surge of AI in drug discovery over the past decade, which is still gaining
popularity. Nonetheless, there are still challenges to be addressed. Despite the prosperity
of deep learning models, it should be emphasized that data is at the core of developing
and evaluating the models. 239,240 To make the models (either predictive or generative) more
useful, data must be in sufficient amount and should maintain high quality. However, a fact
to our dismay is that although existing chemical libraries have a large amount of molecules,
the number of data points for each specific assay can be very scarce. 239 Sometimes, even the
quality of benchmark datasets is questionable with regard to the representative power for
real-world drug discovery imposed by the vast chemical space. 55 Datasets in drug discovery
can be highly imbalanced. 58 Thus, when evaluating the models, there is need to obtain
appropriate datasets and also consider data balancing methods as well as proper evaluation
metrics (e.g., AUPRC vs AUROC). 9
Besides, for the DMTA cycle (see Section “Drug Discovery Overview”) in drug design,
it should always be driven by a need or certain hypotheses. 9 Even equipped with perfect

35
predictive and generative models, a question still remains, i.e., what are the hypotheses
for designing a drug candidate? In other words, what are the desired properties underly-
ing an ideal drug candidate? To generate the insights into drug design, real-world data
(e.g., electronic health records (EHR) and marketed drug databases) 241 is receiving substan-
tial attention for understanding the effectiveness and side effects of different therapeutics.
Recently, we mined a large-scale EHR database for the innate properties underlying opi-
oid analgesics with reduced overdose effects. 242 We also mined the DrugBank database to
identify key pharmacological components (i.e., carriers, transporters, enzymes and targets)
underlying drug-drug interactions (DDIs). 243 These patterns emerging from real-world data
(RWD) allows hypotheses generation and can calibrate drug design insights. 241
Another challenge is that deep learning, despite its superior performance, still leaves the
model elusive for human interpretation. An ongoing need is, therefore, to develop explain-
able models with high interpretability. More specifically, there are four aspects to cover: 31 1)
Transparency, which is knowing how the system reaches a particular answer;2) Justification,
which is elucidating why the answer provided by the model is acceptable;3) Informativeness,
which is providing new information to human decision makers; and 4) Uncertainty estima-
tion, which is quantifying how reliable a prediction is. An ideal state is that AI can allow
scientists to hone their knowledge and beliefs on the investigated process. For more details
on explainable AI in drug discovery, we refer the readers to the review by Jimenez-Luna et
al. 31
In addition to the scientific challenges, technical concerns remain. One unignorable re-
ality is that, even for the state-of-the-art representation learning on molecular graphs, fixed
fingerprints can still outperform GNN-derived representations for molecular property predic-
tion. 244 In fact, ECFPs are a component of some GNN models. 63,71 Besides, there is a lack
of a unified protocol for AI-driven drug discovery studies. For example, different benchmark
datasets, different split folds and evaluation metrics are used across the studies for molecular
property prediction, let alone the varying hyper-parameters tuning, training and evaluation

36
procedure. 244 For molecule generation, Walters et al 239 already proposed a few guidelines to
evaluate the novelty of AI-discovered molecules. Likewise, protocols for molecular property
prediction are also needed.
Overall, there are many promising opportunities as well as significant challenges when
applying AI in drug discovery. In order to launch successful applications, we need to un-
derstand the basic concepts and consider the task, the data, the molecule representation,
the model architecture and the learning paradigm as a whole. In this survey, we have cov-
ered multiple aspects centered around AI-driven drug discovery. We envision that with
these aspects well understood, more meaningful contributions will be made to substantially
transform this field.

Author contributions statement

J.D. and Z.Y. conceived the manuscript and the GitHub repository. J.D. drafted the
manuscript and built the GitHub repository. All authors made critical revisions and re-
view on the manuscript.

Acknowledgement

This project is partially funded by a Stony Brook University OVPR Seed Grant. The neural
networks templates are from Visuals by dair.ai (https://github.com/dair-ai/ml-visuals).

Supporting Information Available

https://github.com/dengjianyuan/Survey AI Drug Discovery

37
References

(1) Mullard, A. New drugs cost US $2.6 billion to develop. Nat. Rev. Drug Discov. 2014,
13, 877.

(2) Dowden, H.; Munro, J. Trends in clinical success rates and therapeutic focus. Nat.
Rev. Drug Discov. 2019, 18, 495–497.

(3) Schneider, G. Automating drug discovery. Nat. Rev. Drug Discov. 2018, 17, 97.

(4) Chen, H.; Engkvist, O.; Wang, Y.; Olivecrona, M.; Blaschke, T. The rise of deep
learning in drug discovery. Drug Discov. Today 2018, 23, 1241–1250.

(5) Mater, A. C.; Coote, M. L. Deep learning in chemistry. J. Chem. Inf. Model 2019,
59, 2545–2559.

(6) Vamathevan, J.; Clark, D.; Czodrowski, P.; Dunham, I.; Ferran, E.; Lee, G.; Li, B.;
Madabhushi, A.; Shah, P.; Spitzer, M., et al. Applications of machine learning in drug
discovery and development. Nat. Rev. Drug Discov. 2019, 18, 463–477.

(7) Paul, D.; Sanap, G.; Shenoy, S.; Kalyane, D.; Kalia, K.; Tekade, R. K. Artificial
intelligence in drug discovery and development. Drug Discovery Today 2021, 26, 80.

(8) Stumpfe, D.; Bajorath, J. Current trends, overlooked issues, and unmet challenges in
virtual screening. J. Chem. Inf. Model 2020, 60, 4112–4115.

(9) Schneider, P.; Walters, W. P.; Plowright, A. T.; Sieroka, N.; Listgarten, J.; Good-
now, R. A.; Fisher, J.; Jansen, J. M.; Duca, J. S.; Rush, T. S., et al. Rethinking drug
design in the artificial intelligence era. Nat. Rev. Drug Discov. 2020, 19, 353–364.

(10) Boström, J.; Brown, D. G.; Young, R. J.; Keserü, G. M. Expanding the medicinal
chemistry synthetic toolbox. Nat. Rev. Drug Discov. 2018, 17, 709–727.

38
(11) Strokach, A.; Becerra, D.; Corbi-Verge, C.; Perez-Riba, A.; Kim, P. M. Fast and
flexible protein design using deep graph neural networks. Cell Syst. 2020, 11, 402–
411.

(12) Pushpakom, S.; Iorio, F.; Eyers, P. A.; Escott, K. J.; Hopper, S.; Wells, A.; Doig, A.;
Guilliams, T.; Latimer, J.; McNamee, C., et al. Drug repurposing: progress, challenges
and recommendations. Nat. Rev. Drug Discov. 2019, 18, 41–58.

(13) Tsigelny, I. F. Artificial intelligence in drug combination therapy. Brief. Bioinformatics


2019, 20, 1434–1448.

(14) Paananen, J.; Fortino, V. An omics perspective on drug target discovery platforms.
Brief. Bioinformatics 2020, 21, 1937–1953.

(15) Hughes, J. P.; Rees, S.; Kalindjian, S. B.; Philpott, K. L. Principles of early drug
discovery. Br. J. Pharmacol. 2011, 162, 1239–1249.

(16) Pereira, D.; Williams, J. Origin and evolution of high throughput screening. Br. J.
Pharmacol. 2007, 152, 53–61.

(17) Bender, A.; Bojanic, D.; Davies, J. W.; Crisman, T. J.; Mikhailov, D.; Scheiber, J.;
Jenkins, J. L.; Deng, Z.; Hill, W. A. G.; Popov, M., et al. Which aspects of HTS are
empirically correlated with downstream success? Curr Opin Drug Discov Devel 2008,
11, 327.

(18) Wang, Y.; Bryant, S. H.; Cheng, T.; Wang, J.; Gindulyte, A.; Shoemaker, B. A.;
Thiessen, P. A.; He, S.; Zhang, J. Pubchem bioassay: 2017 update. Nucleic Acids Res.
2017, 45, D955–D963.

(19) Sterling, T.; Irwin, J. J. ZINC 15–ligand discovery for everyone. J. Chem. Inf. Model
2015, 55, 2324–2337.

39
(20) Kim, S. Getting the most out of PubChem for virtual screening. Expert Opin Drug
Discov 2016, 11, 843–855.

(21) Scior, T.; Bender, A.; Tresadern, G.; Medina-Franco, J. L.; Martı́nez-Mayorga, K.;
Langer, T.; Cuanalo-Contreras, K.; Agrafiotis, D. K. Recognizing pitfalls in virtual
screening: a critical review. J. Chem. Inf. Model 2012, 52, 867–881.

(22) Salahudeen, M. S.; Nishtala, P. S. An overview of pharmacodynamic modelling, ligand-


binding approach and its application in clinical practice. Saudi Pharm J 2017, 25,
165–175.

(23) Hu, Y.; Bajorath, J. Compound promiscuity: what can we learn from current data?
Drug Discov. Today 2013, 18, 644–650.

(24) Yusof, I.; Shah, F.; Hashimoto, T.; Segall, M. D.; Greene, N. Finding the rules for
successful drug optimisation. Drug Discov. Today 2014, 19, 680–687.

(25) Nicolaou, C. A.; Brown, N. Multi-objective optimization methods in drug design. Drug
Discov. Today: Technologies 2013, 10, e427–e435.

(26) Muratov, E. N.; Bajorath, J.; Sheridan, R. P.; Tetko, I. V.; Filimonov, D.; Poroikov, V.;
Oprea, T. I.; Baskin, I. I.; Varnek, A.; Roitberg, A., et al. QSAR without borders.
Chem. Soc. Rev. 2020, 49, 3525–3564.

(27) Schneider, G.; Fechner, U. Computer-based de novo design of drug-like molecules. Nat.
Rev. Drug Discov. 2005, 4, 649–663.

(28) Dobson, C. M. Chemical space and biology. 2004.

(29) Sliwoski, G.; Kothiwale, S.; Meiler, J.; Lowe, E. W. Computational methods in drug
discovery. Pharmacol. Rev. 2014, 66, 334–395.

(30) Van Drie, J. H. Computer-aided drug design: the next 20 years. J. Comput. Aided
Mol. Des. 2007, 21, 591–601.

40
(31) Jiménez-Luna, J.; Grisoni, F.; Schneider, G. Drug discovery with explainable artificial
intelligence. Nat. Mach. Intell. 2020, 2, 573–584.

(32) Bajorath, J. Integration of virtual and high-throughput screening. Nat. Rev. Drug
Discov. 2002, 1, 882–894.

(33) Schneider, G. Virtual screening: an endless staircase? Nat. Rev. Drug Discov. 2010,
9, 273–276.

(34) Polishchuk, P. Interpretation of quantitative structure–activity relationship models:


past, present, and future. J. Chem. Inf. Model 2017, 57, 2618–2639.

(35) Sydow, D.; Burggraaff, L.; Szengel, A.; van Vlijmen, H. W.; IJzerman, A. P.; van
Westen, G. J.; Volkamer, A. Advances and challenges in computational target predic-
tion. J. Chem. Inf. Model 2019, 59, 1728–1742.

(36) Maggiora, G. On outliers and activity cliffs–why QSAR often disappoints. J Chem Inf
Model 2006, 46, 1535–1535.

(37) Stumpfe, D.; Hu, Y.; Dimova, D.; Bajorath, J. Recent progress in understanding
activity cliffs and their utility in medicinal chemistry: miniperspective. J. Med. Chem.
2014, 57, 18–28.

(38) Bajorath, J. Duality of activity cliffs in drug discovery. Expert Opin Drug Discov 2019,
14, 517–520.

(39) Ma, J.; Sheridan, R. P.; Liaw, A.; Dahl, G. E.; Svetnik, V. Deep neural nets as a
method for quantitative structure–activity relationships. J. Chem. Inf. Model 2015,
55, 263–274.

(40) Lavecchia, A. Machine-learning approaches in drug discovery: methods and applica-


tions. Drug Discov. Today 2015, 20, 318–331.

41
(41) Krizhevsky, A.; Sutskever, I.; Hinton, G. E. Imagenet classification with deep convo-
lutional neural networks. Advances in Neural Information Processing Systems 2012,
25, 1097–1105.

(42) Alom, M. Z.; Taha, T. M.; Yakopcic, C.; Westberg, S.; Sidike, P.; Nasrin, M. S.;
Van Esesn, B. C.; Awwal, A. A. S.; Asari, V. K. The history began from alexnet: A
comprehensive survey on deep learning approaches. arXiv preprint arXiv:1803.01164
2018,

(43) Öztürk, H.; Özgür, A.; Schwaller, P.; Laino, T.; Ozkirimli, E. Exploring chemical space
using natural language processing methodologies for drug discovery. Drug Discov.
Today 2020, 25, 689–705.

(44) Jiménez-Luna, J.; Grisoni, F.; Weskamp, N.; Schneider, G. Artificial intelligence in
drug discovery: Recent advances and future perspectives. Expert Opin Drug Discov
2021, 1–11.

(45) Zhavoronkov, A.; Ivanenkov, Y. A.; Aliper, A.; Veselov, M. S.; Aladinskiy, V. A.;
Aladinskaya, A. V.; Terentiev, V. A.; Polykovskiy, D. A.; Kuznetsov, M. D.; Asadu-
laev, A., et al. Deep learning enables rapid identification of potent DDR1 kinase
inhibitors. Nat. Biotechnol. 2019, 37, 1038–1040.

(46) Stokes, J. M.; Yang, K.; Swanson, K.; Jin, W.; Cubillos-Ruiz, A.; Donghia, N. M.;
MacNair, C. R.; French, S.; Carfrae, L. A.; Bloom-Ackermann, Z., et al. A deep
learning approach to antibiotic discovery. Cell 2020, 180, 688–702.

(47) Chuang, K. V.; Gunsalus, L. M.; Keiser, M. J. Learning Molecular Representations


for Medicinal Chemistry: Miniperspective. J. Med. Chem. 2020, 63, 8705–8722.

(48) Mayr, A.; Klambauer, G.; Unterthiner, T.; Hochreiter, S. DeepTox: toxicity prediction
using deep learning. Frontiers in Environmental Science 2016, 3, 80.

42
(49) Andrade, R. J.; Chalasani, N.; Björnsson, E. S.; Suzuki, A.; Kullak-Ublick, G. A.;
Watkins, P. B.; Devarbhavi, H.; Merz, M.; Lucena, M. I.; Kaplowitz, N., et al. Drug-
induced liver injury. Nat. Rev. Dis. Primers 2019, 5, 1–22.

(50) Elton, D. C.; Boukouvalas, Z.; Fuge, M. D.; Chung, P. W. Deep learning for molecular
design—a review of the state of the art. Mol. Syst. Des. Eng. 2019, 4, 828–849.

(51) Mercado, R.; Rastemo, T.; Lindelöf, E.; Klambauer, G.; Engkvist, O.; Chen, H.; Bjer-
rum, E. J. Practical Notes on Building Molecular Graph Generative Models. Applied
AI Letters 2020,

(52) Schaduangrat, N.; Lampa, S.; Simeon, S.; Gleeson, M. P.; Spjuth, O.; Nantase-
namat, C. Towards reproducible computational drug discovery. J. Cheminformatics
2020, 12, 9.

(53) Bender, A.; Cortes-Ciriano, I. Artificial intelligence in drug discovery: what is realistic,
what are illusions? Part 1: Ways to make an impact, and why we are not there yet.
Drug Discov. Today 2020,

(54) Bender, A.; Cortes-Ciriano, I. Artificial intelligence in drug discovery: what is realistic,
what are illusions? Part 2: a discussion of chemical and biological data used for AI in
drug discovery. Drug Discov. Today 2021,

(55) Walters, W. P.; Barzilay, R. Critical assessment of AI in drug discovery. Expert Opin
Drug Discov 2021, 1–11.

(56) Rifaioglu, A. S.; Atas, H.; Martin, M. J.; Cetin-Atalay, R.; Atalay, V.; Doğan, T. Re-
cent applications of deep learning and machine intelligence on in silico drug discovery:
methods, tools and databases. Brief. Bioinformatics 2019, 20, 1878–1912.

(57) Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B. A.;

43
Thiessen, P. A.; Yu, B., et al. PubChem in 2021: new data content and improved web
interfaces. Nucleic Acids Res. 2021, 49, D1388–D1395.

(58) Korkmaz, S. Deep learning-based imbalanced data classification for drug discovery. J.
Chem. Inf. Model 2020, 60, 4180–4190.

(59) Chithrananda, S.; Grand, G.; Ramsundar, B. ChemBERTa: Large-Scale


Self-Supervised Pretraining for Molecular Property Prediction. arXiv preprint
arXiv:2010.09885 2020,

(60) Gaulton, A.; Hersey, A.; Nowotka, M.; Bento, A. P.; Chambers, J.; Mendez, D.; Mu-
towo, P.; Atkinson, F.; Bellis, L. J.; Cibrián-Uhalte, E., et al. The ChEMBL database
in 2017. Nucleic Acids Res. 2017, 45, D945–D954.

(61) Davies, M.; Nowotka, M.; Papadatos, G.; Dedman, N.; Gaulton, A.; Atkinson, F.; Bel-
lis, L.; Overington, J. P. ChEMBL web services: streamlining access to drug discovery
data and utilities. Nucleic Acids Res. 2015, 43, W612–W620.

(62) Mayr, A.; Klambauer, G.; Unterthiner, T.; Steijaert, M.; Wegner, J. K.; Ceule-
mans, H.; Clevert, D.-A.; Hochreiter, S. Large-scale comparison of machine learning
methods for drug target prediction on ChEMBL. Chem. Sci 2018, 9, 5441–5451.

(63) Rong, Y.; Bian, Y.; Xu, T.; Xie, W.; Wei, Y.; Huang, W.; Huang, J. Grover: Self-
supervised message passing transformer on large-scale molecular data. arXiv preprint
arXiv:2007.02835 2020,

(64) Polykovskiy, D.; Zhebrak, A.; Sanchez-Lengeling, B.; Golovanov, S.; Tatanov, O.;
Belyaev, S.; Kurbanov, R.; Artamonov, A.; Aladinskiy, V.; Veselov, M., et al. Molec-
ular sets (MOSES): a benchmarking platform for molecular generation models. Front.
Pharmacol. 2020, 11 .

44
(65) Lagarde, N.; Zagury, J.-F.; Montes, M. Benchmarking data sets for the evaluation of
virtual ligand screening methods: review and perspectives. J. Chem. Inf. Model 2015,
55, 1297–1307.

(66) Chen, M.; Suzuki, A.; Thakkar, S.; Yu, K.; Hu, C.; Tong, W. DILIrank: the largest ref-
erence drug list ranked by the risk for developing drug-induced liver injury in humans.
Drug Discov Today 2016, 21, 648–653.

(67) David, L.; Thakkar, A.; Mercado, R.; Engkvist, O. Molecular representations in AI-
driven drug discovery: a review and practical guide. J. Cheminformatics 2020, 12,
1–22.

(68) Morgan, H. L. The generation of a unique machine description for chemical structures-
a technique developed at chemical abstracts service. J. Chem. Doc 1965, 5, 107–113.

(69) Subramanian, G.; Ramsundar, B.; Pande, V.; Denny, R. A. Computational modeling
of β-secretase 1 (BACE-1) inhibitors using ligand based approaches. J. Chem. Inf.
Model 2016, 56, 1936–1949.

(70) Zang, Q.; Mansouri, K.; Williams, A. J.; Judson, R. S.; Allen, D. G.; Casey, W. M.;
Kleinstreuer, N. C. In silico prediction of physicochemical properties of environmental
chemicals using molecular fingerprints and machine learning. J. Chem. Inf. Model
2017, 57, 36–49.

(71) Yang, K.; Swanson, K.; Jin, W.; Coley, C.; Eiden, P.; Gao, H.; Guzman-Perez, A.;
Hopper, T.; Kelley, B.; Mathea, M., et al. Analyzing learned molecular representations
for property prediction. J. Chem. Inf. Model 2019, 59, 3370–3388.

(72) Mercado, R.; Rastemo, T.; Lindelöf, E.; Klambauer, G.; Engkvist, O.; Chen, H.;
Bjerrum, E. J. Graph Networks for Molecular Design. Mach. Learn.: Sci. Technol.
2020,

45
(73) Jin, W.; Barzilay, R.; Jaakkola, T. Multi-objective molecule generation using inter-
pretable substructures. International Conference on Machine Learning. 2020; pp 4849–
4859.

(74) Weininger, D. SMILES, a chemical language and information system. 1. Introduction


to methodology and encoding rules. J Chem Inform Comput Sci 1988, 28, 31–36.

(75) Weininger, D.; Weininger, A.; Weininger, J. L. SMILES. 2. Algorithm for generation
of unique SMILES notation. J Chem Inform Comput Sci 1989, 29, 97–101.

(76) Bian, Y.; Xie, X.-Q. Generative chemistry: drug discovery with deep learning gener-
ative models. J. Mol. Model. 2021, 27, 1–18.

(77) Xiong, Z.; Wang, D.; Liu, X.; Zhong, F.; Wan, X.; Li, X.; Li, Z.; Luo, X.; Chen, K.;
Jiang, H., et al. Pushing the boundaries of molecular representation for drug discovery
with the graph attention mechanism. J. Med. Chem. 2019, 63, 8749–8760.

(78) Gómez-Bombarelli, R.; Wei, J. N.; Duvenaud, D.; Hernández-Lobato, J. M.; Sánchez-
Lengeling, B.; Sheberla, D.; Aguilera-Iparraguirre, J.; Hirzel, T. D.; Adams, R. P.;
Aspuru-Guzik, A. Automatic chemical design using a data-driven continuous repre-
sentation of molecules. ACS Cent. Sci. 2018, 4, 268–276.

(79) Popova, M.; Isayev, O.; Tropsha, A. Deep reinforcement learning for de novo drug
design. Sci. Adv. 2018, 4, eaap7885.

(80) Ragoza, M.; Hochuli, J.; Idrobo, E.; Sunseri, J.; Koes, D. R. Protein–ligand scoring
with convolutional neural networks. J. Chem. Inf. Model 2017, 57, 942–957.

(81) Jiménez, J.; Skalic, M.; Martinez-Rosell, G.; De Fabritiis, G. K deep: protein–ligand
absolute binding affinity prediction via 3d-convolutional neural networks. J. Chem.
Inf. Model 2018, 58, 287–296.

46
(82) Lim, J.; Ryu, S.; Park, K.; Choe, Y. J.; Ham, J.; Kim, W. Y. Predicting drug–target
interaction using a novel graph neural network with 3D structure-embedded graph
representation. J. Chem. Inf. Model 2019, 59, 3981–3988.

(83) Hernandez, M.; Liang Gan, G.; Linvill, K.; Dukatz, C.; Feng, J.; Bhisetti, G. A
quantum-inspired method for three-dimensional ligand-based virtual screening. J.
Chem. Inf. Model 2019, 59, 4475–4485.

(84) Wu, K.; Wei, G.-W. Quantitative toxicity prediction using topology based multitask
deep neural networks. J. Chem. Inf. Model 2018, 58, 520–531.

(85) Skalic, M.; Jiménez, J.; Sabbadin, D.; De Fabritiis, G. Shape-based generative model-
ing for de novo drug design. J. Chem. Inf. Model 2019, 59, 1205–1214.

(86) Simm, G.; Pinsler, R.; Hernández-Lobato, J. M. Reinforcement learning for molecular
design guided by quantum mechanics. International Conference on Machine Learning.
2020; pp 8959–8969.

(87) Hemmerich, J.; Asilar, E.; Ecker, G. F. COVER: conformational oversampling as data
augmentation for molecules. J. Cheminformatics 2020, 12, 1–12.

(88) Fernandez, M.; Ban, F.; Woo, G.; Hsing, M.; Yamazaki, T.; LeBlanc, E.; Rennie, P. S.;
Welch, W. J.; Cherkasov, A. Toxic colors: the use of deep learning for predicting
toxicity of compounds merely from their graphic images. J. Chem. Inf. Model 2018,
58, 1533–1543.

(89) Meyer, J. G.; Liu, S.; Miller, I. J.; Coon, J. J.; Gitter, A. Learning drug functions
from chemical structures with convolutional neural networks and random forests. J.
Chem. Inf. Model 2019, 59, 4438–4449.

(90) Cortés-Ciriano, I.; Bender, A. KekuleScope: prediction of cancer cell line sensitivity

47
and compound potency using convolutional neural networks trained on compound
images. J. Cheminformatics 2019, 11, 1–16.

(91) Rifaioglu, A. S.; Nalbat, E.; Atalay, V.; Martin, M. J.; Cetin-Atalay, R.; Doğan, T.
DEEPScreen: high performance drug–target interaction prediction with convolutional
neural networks using 2-D structural compound representations. Chem. Sci 2020, 11,
2531–2557.

(92) Rajan, K.; Brinkhaus, H. O.; Sorokina, M.; Zielesny, A.; Steinbeck, C. DECIMER-
Segmentation: Automated extraction of chemical structure depictions from scientific
literature. J. Cheminformatics 2021, 13, 1–9.

(93) Wu, Z.; Ramsundar, B.; Feinberg, E. N.; Gomes, J.; Geniesse, C.; Pappu, A. S.;
Leswing, K.; Pande, V. MoleculeNet: a benchmark for molecular machine learning.
Chem. Sci 2018, 9, 513–530.

(94) Ramsundar, B.; Eastman, P.; Walters, P.; Pande, V.; Leswing, K.; Wu, Z. Deep
Learning for the Life Sciences; O’Reilly Media, 2019; https://www.amazon.com/
Deep-Learning-Life-Sciences-Microscopy/dp/1492039837.

(95) Fabian, B.; Edlich, T.; Gaspar, H.; Segler, M.; Meyers, J.; Fiscato, M.; Ahmed, M.
Molecular representation learning with language models and domain-relevant auxiliary
tasks. arXiv preprint arXiv:2011.13230 2020,

(96) Shen, W. X.; Zeng, X.; Zhu, F.; li Wang, Y.; Qin, C.; Tan, Y.; Jiang, Y. Y.;
Chen, Y. Z. Out-of-the-box deep learning prediction of pharmaceutical properties by
broadly learned knowledge-based molecular representations. Nat. Mach. Intell. 2021,
3, 334–343.

(97) Olivecrona, M.; Blaschke, T.; Engkvist, O.; Chen, H. Molecular de-novo design
through deep reinforcement learning. J. Cheminformatics 2017, 9, 1–14.

48
(98) Blaschke, T.; Arús-Pous, J.; Chen, H.; Margreitter, C.; Tyrchan, C.; Engkvist, O.;
Papadopoulos, K.; Patronov, A. REINVENT 2.0: An AI Tool for De Novo Drug
Design. J. Chem. Inf. Model 2020,

(99) Brown, N.; Fiscato, M.; Segler, M. H.; Vaucher, A. C. GuacaMol: benchmarking
models for de novo molecular design. J. Chem. Inf. Model 2019, 59, 1096–1108.

(100) Segler, M. H.; Kogej, T.; Tyrchan, C.; Waller, M. P. Generating focused molecule
libraries for drug discovery with recurrent neural networks. ACS Cent. Sci. 2018, 4,
120–131.

(101) Walters, W. P.; Murcko, M. A. Prediction of ‘drug-likeness’. Adv. Drug Deliv. Rev.
2002, 54, 255–271.

(102) Schneider, N.; Jäckels, C.; Andres, C.; Hutter, M. C. Gradual in silico filtering for
druglike substances. J. Chem. Inf. Model 2008, 48, 613–628.

(103) Palmer, D. S.; O’Boyle, N. M.; Glen, R. C.; Mitchell, J. B. Random forest models to
predict aqueous solubility. J. Chem. Inf. Model 2007, 47, 150–158.

(104) Schroeter, T.; Schwaighofer, A.; Mika, S.; Ter Laak, A.; Suelzle, D.; Ganzer, U.;
Heinrich, N.; Müller, K.-R. Machine learning models for lipophilicity and their domain
of applicability. Mol. Pharm. 2007, 4, 524–538.

(105) Hou, T.; Wang, J.; Li, Y. ADME evaluation in drug discovery. 8. The prediction of
human intestinal absorption by a support vector machine. J. Chem. Inf. Model 2007,
47, 2408–2415.

(106) Tian, S.; Li, Y.; Wang, J.; Zhang, J.; Hou, T. ADME evaluation in drug discovery.
9. Prediction of oral bioavailability in humans based on molecular properties and
structural fingerprints. Mol. Pharm. 2011, 8, 841–851.

49
(107) Sakiyama, Y.; Yuki, H.; Moriya, T.; Hattori, K.; Suzuki, M.; Shimada, K.; Honma, T.
Predicting human liver microsomal stability with machine learning techniques. J. Mol.
Graph. Model. 2008, 26, 907–915.

(108) Vasanthanathan, P.; Taboureau, O.; Oostenbrink, C.; Vermeulen, N. P.; Olsen, L.;
Jørgensen, F. S. Classification of cytochrome P450 1A2 inhibitors and noninhibitors
by machine learning techniques. Drug Metab. Dispos 2009, 37, 658–664.

(109) Riddick, G.; Song, H.; Ahn, S.; Walling, J.; Borges-Rivera, D.; Zhang, W.; Fine, H. A.
Predicting in vitro drug sensitivity using Random Forests. Bioinformatics 2011, 27,
220–224.

(110) Zhao, C.; Zhang, H.; Zhang, X.; Liu, M.; Hu, Z.; Fan, B. Application of support vector
machine (SVM) for prediction toxic activity of different data sets. Toxicology 2006,
217, 105–119.

(111) Heikamp, K.; Bajorath, J. Support vector machines for drug discovery. Expert Opin
Drug Discov 2014, 9, 93–104.

(112) Svetnik, V.; Liaw, A.; Tong, C.; Culberson, J. C.; Sheridan, R. P.; Feuston, B. P.
Random forest: a classification and regression tool for compound classification and
QSAR modeling. J Chem Inform Comput Sci 2003, 43, 1947–1958.

(113) Dahl, G. Deep learning how I did it: Merck 1st place interview. Online article available
from http://blog. kaggle. com/2012/11/01/deep-learning-how-i-did-it-merck-1st-place-
interview 2012,

(114) LeCun, Y.; Bengio, Y.; Hinton, G. Deep learning. Nature 2015, 521, 436–444.

(115) Simm, J.; Klambauer, G.; Arany, A.; Steijaert, M.; Wegner, J. K.; Gustin, E.;
Chupakhin, V.; Chong, Y. T.; Vialard, J.; Buijnsters, P., et al. Repurposing high-

50
throughput image assays enables biological activity prediction for drug discovery. Cell
Chem. Biol. 2018, 25, 611–618.

(116) Hofmarcher, M.; Rumetshofer, E.; Clevert, D.-A.; Hochreiter, S.; Klambauer, G. Ac-
curate prediction of biological assays with high-throughput microscopy images and
convolutional networks. J. Chem. Inf. Model 2019, 59, 1163–1171.

(117) Ramsundar, B.; Kearnes, S.; Riley, P.; Webster, D.; Konerding, D.; Pande, V. Mas-
sively multitask networks for drug discovery. arXiv preprint arXiv:1502.02072 2015,

(118) Duvenaud, D.; Maclaurin, D.; Aguilera-Iparraguirre, J.; Gómez-Bombarelli, R.;


Hirzel, T.; Aspuru-Guzik, A.; Adams, R. P. Convolutional networks on graphs for
learning molecular fingerprints. arXiv preprint arXiv:1509.09292 2015,

(119) Glen, R. C.; Bender, A.; Arnby, C. H.; Carlsson, L.; Boyer, S.; Smith, J. Circular
fingerprints: flexible molecular descriptors with applications from physical chemistry
to ADME. IDrugs 2006, 9, 199.

(120) Goh, G. B.; Siegel, C.; Vishnu, A.; Hodas, N. O.; Baker, N. Chemception: a deep
neural network with minimal chemistry knowledge matches the performance of expert-
developed QSAR/QSPR models. arXiv preprint arXiv:1706.06689 2017,

(121) Huang, G.; Liu, Z.; Van Der Maaten, L.; Weinberger, K. Q. Densely connected con-
volutional networks. Proceedings of the IEEE Conference on Computer Vision and
Pattern Recognition. 2017; pp 4700–4708.

(122) He, K.; Zhang, X.; Ren, S.; Sun, J. Deep residual learning for image recognition.
Proceedings of the IEEE Conference on Computer Vision and Pattern Recognition.
2016; pp 770–778.

(123) Simonyan, K.; Zisserman, A. Very deep convolutional networks for large-scale image
recognition. arXiv preprint arXiv:1409.1556 2014,

51
(124) Staker, J.; Marshall, K.; Abel, R.; McQuaw, C. M. Molecular structure extraction
from documents using deep learning. J. Chem. Inf. Model 2019, 59, 1017–1029.

(125) Rajan, K.; Zielesny, A.; Steinbeck, C. DECIMER: towards deep learning for chemical
image recognition. J. Cheminformatics 2020, 12, 1–9.

(126) Hossain, M. Z.; Sohel, F.; Shiratuddin, M. F.; Laga, H. A comprehensive survey of
deep learning for image captioning. ACM Computing Surveys (CsUR) 2019, 51, 1–36.

(127) Mikolov, T.; Kombrink, S.; Burget, L.; Černockỳ, J.; Khudanpur, S. Extensions of
recurrent neural network language model. 2011 IEEE International Conference on
Acoustics, Speech and Signal Processing (ICASSP). 2011; pp 5528–5531.

(128) Boulanger-Lewandowski, N.; Bengio, Y.; Vincent, P. Modeling temporal dependen-


cies in high-dimensional sequences: Application to polyphonic music generation and
transcription. arXiv preprint arXiv:1206.6392 2012,

(129) Hochreiter, S.; Schmidhuber, J. Long short-term memory. Neural Comput. 1997, 9,
1735–1780.

(130) Chung, J.; Gulcehre, C.; Cho, K.; Bengio, Y. Empirical evaluation of gated recurrent
neural networks on sequence modeling. arXiv preprint arXiv:1412.3555 2014,

(131) Goh, G. B.; Hodas, N. O.; Siegel, C.; Vishnu, A. Smiles2vec: An interpretable
general-purpose deep neural network for predicting chemical properties. arXiv preprint
arXiv:1712.02034 2017,

(132) Neil, D.; Segler, M.; Guasch, L.; Ahmed, M.; Plumbley, D.; Sellwood, M.; Brown, N.
Exploring deep recurrent models with reinforcement learning for molecule design. Pro-
ceedings of The International Conference on Learning Representations. 2018.

(133) Joulin, A.; Mikolov, T. Inferring algorithmic patterns with stack-augmented recurrent
nets. arXiv preprint arXiv:1503.01007 2015,

52
(134) Ståhl, N.; Falkman, G.; Karlsson, A.; Mathiason, G.; Bostrom, J. Deep reinforcement
learning for multiparameter optimization in de novo drug design. J. Chem. Inf. Model
2019, 59, 3166–3176.

(135) Zheng, S.; Yan, X.; Yang, Y.; Xu, J. Identifying structure–property relationships
through SMILES syntax analysis with self-attention mechanism. J. Chem. Inf. Model
2019, 59, 914–923.

(136) You, J.; Ying, R.; Ren, X.; Hamilton, W.; Leskovec, J. Graphrnn: Generating real-
istic graphs with deep auto-regressive models. International Conference on Machine
Learning. 2018; pp 5708–5717.

(137) Li, Y.; Vinyals, O.; Dyer, C.; Pascanu, R.; Battaglia, P. Learning deep generative
models of graphs. arXiv preprint arXiv:1803.03324 2018,

(138) Li, Y.; Zhang, L.; Liu, Z. Multi-objective de novo drug design with conditional graph
generative model. J. Cheminformatics 2018, 10, 1–24.

(139) Popova, M.; Shvets, M.; Oliva, J.; Isayev, O. MolecularRNN: Generating realistic
molecular graphs with optimized properties. arXiv preprint arXiv:1905.13372 2019,

(140) You, J.; Liu, B.; Ying, R.; Pande, V.; Leskovec, J. Graph convolutional policy network
for goal-directed molecular graph generation. arXiv preprint arXiv:1806.02473 2018,

(141) Sattarov, B.; Baskin, I. I.; Horvath, D.; Marcou, G.; Bjerrum, E. J.; Varnek, A. De
novo molecular design by combining deep autoencoder recurrent neural networks with
generative topographic mapping. J. Chem. Inf. Model 2019, 59, 1182–1196.

(142) Guimaraes, G. L.; Sanchez-Lengeling, B.; Outeiral, C.; Farias, P. L. C.; Aspuru-
Guzik, A. Objective-reinforced generative adversarial networks (ORGAN) for sequence
generation models. arXiv preprint arXiv:1705.10843 2017,

53
(143) Sanchez-Lengeling, B.; Outeiral, C.; Guimaraes, G. L.; Aspuru-Guzik, A. Optimiz-
ing distributions over molecular space. An objective-reinforced generative adversarial
network for inverse-design chemistry (ORGANIC). ChemRxiv 2017, 2017 .

(144) Wu, Z.; Pan, S.; Chen, F.; Long, G.; Zhang, C.; Philip, S. Y. A comprehensive survey
on graph neural networks. IEEE Trans Neural Netw Learn 2020,

(145) Li, Y.; Tarlow, D.; Brockschmidt, M.; Zemel, R. Gated graph sequence neural net-
works. arXiv preprint arXiv:1511.05493 2015,

(146) Defferrard, M.; Bresson, X.; Vandergheynst, P. Convolutional neural networks on


graphs with fast localized spectral filtering. arXiv preprint arXiv:1606.09375 2016,

(147) Kipf, T. N.; Welling, M. Semi-supervised classification with graph convolutional net-
works. arXiv preprint arXiv:1609.02907 2016,

(148) Gilmer, J.; Schoenholz, S. S.; Riley, P. F.; Vinyals, O.; Dahl, G. E. Neural message
passing for quantum chemistry. International Conference on Machine Learning. 2017;
pp 1263–1272.

(149) Hamilton, W. L.; Ying, R.; Leskovec, J. Inductive representation learning on large
graphs. arXiv preprint arXiv:1706.02216 2017,

(150) Veličković, P.; Cucurull, G.; Casanova, A.; Romero, A.; Lio, P.; Bengio, Y. Graph
attention networks. arXiv preprint arXiv:1710.10903 2017,

(151) Xu, K.; Hu, W.; Leskovec, J.; Jegelka, S. How powerful are graph neural networks?
arXiv preprint arXiv:1810.00826 2018,

(152) Kearnes, S.; McCloskey, K.; Berndl, M.; Pande, V.; Riley, P. Molecular graph convo-
lutions: moving beyond fingerprints. J. Comput. Aided Mol. Des. 2016, 30, 595–608.

(153) Landrum, G. RDKit: Open-Source Cheminformatics Software. RDKit 2016,

54
(154) Withnall, M.; Lindelöf, E.; Engkvist, O.; Chen, H. Building attention and edge mes-
sage passing neural networks for bioactivity and physical–chemical property predic-
tion. J. Cheminformatics 2020, 12, 1–18.

(155) Schütt, K. T.; Kindermans, P.-J.; Sauceda, H. E.; Chmiela, S.; Tkatchenko, A.;
Müller, K.-R. Schnet: A continuous-filter convolutional neural network for modeling
quantum interactions. arXiv preprint arXiv:1706.08566 2017,

(156) Feinberg, E. N.; Sur, D.; Wu, Z.; Husic, B. E.; Mai, H.; Li, Y.; Sun, S.; Yang, J.;
Ramsundar, B.; Pande, V. S. PotentialNet for molecular property prediction. ACS
Cent. Sci. 2018, 4, 1520–1530.

(157) Klicpera, J.; Groß, J.; Günnemann, S. Directional message passing for molecular
graphs. arXiv preprint arXiv:2003.03123 2020,

(158) Altae-Tran, H.; Ramsundar, B.; Pappu, A. S.; Pande, V. Low data drug discovery
with one-shot learning. ACS Cent. Sci. 2017, 3, 283–293.

(159) Liu, S.; Demirel, M. F.; Liang, Y. N-gram graph: Simple unsupervised representation
for graphs, with applications to molecules. arXiv preprint arXiv:1806.09206 2018,

(160) Lu, C.; Liu, Q.; Wang, C.; Huang, Z.; Lin, P.; He, L. Molecular property prediction:
A multilevel quantum interactions modeling perspective. Proceedings of the AAAI
Conference on Artificial Intelligence. 2019; pp 1052–1060.

(161) Cai, C.; Guo, P.; Zhou, Y.; Zhou, J.; Wang, Q.; Zhang, F.; Fang, J.; Cheng, F. Deep
learning-based prediction of drug-induced cardiotoxicity. J. Chem. Inf. Model 2019,
59, 1073–1084.

(162) Wang, X.; Li, Z.; Jiang, M.; Wang, S.; Zhang, S.; Wei, Z. Molecule property prediction
based on spatial graph embedding. J. Chem. Inf. Model 2019, 59, 3817–3828.

55
(163) Hu, W.; Liu, B.; Gomes, J.; Zitnik, M.; Liang, P.; Pande, V.; Leskovec, J. Strategies
for pre-training graph neural networks. arXiv preprint arXiv:1905.12265 2019,

(164) Hao, Z.; Lu, C.; Huang, Z.; Wang, H.; Hu, Z.; Liu, Q.; Chen, E.; Lee, C. ASGN: An
active semi-supervised graph neural network for molecular property prediction. Pro-
ceedings of the 26th ACM SIGKDD International Conference on Knowledge Discovery
& Data Mining. 2020; pp 731–752.

(165) Nguyen, C. Q.; Kreatsoulas, C.; Branson, K. M. Meta-Learning Initializations for


Low-Resource Drug Discovery. arXiv preprint arXiv:2003.05996 2020,

(166) Li, X.; Yan, X.; Gu, Q.; Zhou, H.; Wu, D.; Xu, J. DeepChemStable: Chemical stability
prediction with an attention-based graph convolution network. J. Chem. Inf. Model
2019, 59, 1044–1049.

(167) Tang, B.; Kramer, S. T.; Fang, M.; Qiu, Y.; Wu, Z.; Xu, D. A self-attention based
message passing neural network for predicting molecular lipophilicity and aqueous
solubility. J. Cheminformatics 2020, 12, 1–9.

(168) Pathak, Y.; Laghuvarapu, S.; Mehta, S.; Priyakumar, U. D. Chemically interpretable
graph interaction network for prediction of pharmacokinetic properties of drug-like
molecules. Proceedings of the AAAI Conference on Artificial Intelligence. 2020; pp
873–880.

(169) Zhou, Z.; Kearnes, S.; Li, L.; Zare, R. N.; Riley, P. Optimization of molecules via deep
reinforcement learning. Scientific reports 2019, 9, 1–10.

(170) Khemchandani, Y.; O’Hagan, S.; Samanta, S.; Swainston, N.; Roberts, T. J.; Bolle-
gala, D.; Kell, D. B. DeepGraphMolGen, a multi-objective, computational strategy for
generating molecules with desirable properties: a graph convolution and reinforcement
learning approach. J. Cheminformatics 2020, 12, 1–17.

56
(171) Xu, C.; Liu, Q.; Huang, M.; Jiang, T. Reinforced Molecular Optimization with
Neighborhood-Controlled Grammars. arXiv preprint arXiv:2011.07225 2020,

(172) Kingma, D. P.; Welling, M. Auto-encoding variational bayes. arXiv preprint


arXiv:1312.6114 2013,

(173) Kingma, D. P.; Welling, M. An introduction to variational autoencoders. arXiv


preprint arXiv:1906.02691 2019,

(174) Kusner, M. J.; Paige, B.; Hernández-Lobato, J. M. Grammar variational autoencoder.


International Conference on Machine Learning. 2017; pp 1945–1954.

(175) Dai, H.; Tian, Y.; Dai, B.; Skiena, S.; Song, L. Syntax-directed variational autoencoder
for structured data. arXiv preprint arXiv:1802.08786 2018,

(176) Kang, S.; Cho, K. Conditional molecular design with deep generative models. J. Chem.
Inf. Model 2018, 59, 43–52.

(177) Lim, J.; Ryu, S.; Kim, J. W.; Kim, W. Y. Molecular generative model based on
conditional variational autoencoder for de novo molecular design. J. Cheminformatics
2018, 10, 1–9.

(178) Liu, Q.; Allamanis, M.; Brockschmidt, M.; Gaunt, A. L. Constrained graph variational
autoencoders for molecule design. arXiv preprint arXiv:1805.09076 2018,

(179) Samanta, B.; De, A.; Jana, G.; Gómez, V.; Chattaraj, P. K.; Ganguly, N.; Gomez-
Rodriguez, M. Nevae: A deep generative model for molecular graphs. J Mach Learn
Res 2020,

(180) Chenthamarakshan, V.; Das, P.; Padhi, I.; Strobelt, H.; Lim, K. W.; Hoover, B.;
Hoffman, S. C.; Mojsilovic, A. Target-specific and selective drug design for covid-19
using deep generative models. arXiv preprint arXiv:2004.01215 2020,

57
(181) Makhzani, A.; Shlens, J.; Jaitly, N.; Goodfellow, I.; Frey, B. Adversarial autoencoders.
arXiv preprint arXiv:1511.05644 2015,

(182) Kadurin, A.; Nikolenko, S.; Khrabrov, K.; Aliper, A.; Zhavoronkov, A. druGAN: an
advanced generative adversarial autoencoder model for de novo generation of new
molecules with desired molecular properties in silico. Mol. Pharm. 2017, 14, 3098–
3104.

(183) Blaschke, T.; Olivecrona, M.; Engkvist, O.; Bajorath, J.; Chen, H. Application of
generative autoencoder in de novo molecular design. Mol. Inform. 2018, 37, 1700123.

(184) Polykovskiy, D.; Zhebrak, A.; Vetrov, D.; Ivanenkov, Y.; Aladinskiy, V.;
Mamoshina, P.; Bozdaganyan, M.; Aliper, A.; Zhavoronkov, A.; Kadurin, A. En-
tangled conditional adversarial autoencoder for de novo drug discovery. Mol. Pharm.
2018, 15, 4398–4405.

(185) Simonovsky, M.; Komodakis, N. Graphvae: Towards generation of small graphs us-
ing variational autoencoders. International Conference on Artificial Neural Networks.
2018; pp 412–422.

(186) Jin, W.; Barzilay, R.; Jaakkola, T. Junction tree variational autoencoder for molecular
graph generation. International Conference on Machine Learning. 2018; pp 2323–2332.

(187) Ma, T.; Chen, J.; Xiao, C. Constrained generation of semantically valid graphs via
regularizing variational autoencoders. arXiv preprint arXiv:1809.02630 2018,

(188) Kajino, H. Molecular hypergraph grammar with its application to molecular optimiza-
tion. International Conference on Machine Learning. 2019; pp 3183–3191.

(189) Kwon, Y.; Yoo, J.; Choi, Y.-S.; Son, W.-J.; Lee, D.; Kang, S. Efficient learning of
non-autoregressive graph variational autoencoders for molecular graph generation. J.
Cheminformatics 2019, 11, 1–10.

58
(190) Lim, J.; Hwang, S.-Y.; Kim, S.; Moon, S.; Kim, W. Y. Scaffold-based molecular design
using graph generative model. arXiv preprint arXiv:1905.13639 2019,

(191) Fu, T.; Xiao, C.; Sun, J. Core: Automatic molecule optimization using copy & refine
strategy. Proceedings of the AAAI Conference on Artificial Intelligence. 2020; pp 638–
645.

(192) Kwon, Y.; Lee, D.; Choi, Y.-S.; Shin, K.; Kang, S. Compressed graph representation
for scalable molecular graph generation. J. Cheminformatics 2020, 12, 1–8.

(193) Jin, W.; Barzilay, R.; Jaakkola, T. Hierarchical generation of molecular graphs using
structural motifs. International Conference on Machine Learning. 2020; pp 4839–4848.

(194) Goodfellow, I. J.; Pouget-Abadie, J.; Mirza, M.; Xu, B.; Warde-Farley, D.;
Ozair, S.; Courville, A.; Bengio, Y. Generative adversarial networks. arXiv preprint
arXiv:1406.2661 2014,

(195) Yu, L.; Zhang, W.; Wang, J.; Yu, Y. Seqgan: Sequence generative adversarial nets
with policy gradient. Proceedings of the AAAI Conference on Artificial Intelligence.
2017.

(196) Putin, E.; Asadulaev, A.; Ivanenkov, Y.; Aladinskiy, V.; Sanchez-Lengeling, B.;
Aspuru-Guzik, A.; Zhavoronkov, A. Reinforced adversarial neural computer for de
novo molecular design. J. Chem. Inf. Model 2018, 58, 1194–1204.

(197) De Cao, N.; Kipf, T. MolGAN: An implicit generative model for small molecular
graphs. arXiv preprint arXiv:1805.11973 2018,

(198) Salimans, T.; Goodfellow, I.; Zaremba, W.; Cheung, V.; Radford, A.; Chen, X. Im-
proved techniques for training gans. arXiv preprint arXiv:1606.03498 2016,

(199) Kobyzev, I.; Prince, S.; Brubaker, M. Normalizing flows: An introduction and review
of current methods. IEEE PAMI 2020,

59
(200) Dinh, L.; Krueger, D.; Bengio, Y. Nice: Non-linear independent components estima-
tion. arXiv preprint arXiv:1410.8516 2014,

(201) Dinh, L.; Sohl-Dickstein, J.; Bengio, S. Density estimation using real nvp. arXiv
preprint arXiv:1605.08803 2016,

(202) Madhawa, K.; Ishiguro, K.; Nakago, K.; Abe, M. Graphnvp: An invertible flow model
for generating molecular graphs. arXiv preprint arXiv:1905.11600 2019,

(203) Honda, S.; Akita, H.; Ishiguro, K.; Nakanishi, T.; Oono, K. Graph residual flow for
molecular graph generation. arXiv preprint arXiv:1909.13521 2019,

(204) Shi, C.; Xu, M.; Zhu, Z.; Zhang, W.; Zhang, M.; Tang, J. Graphaf: a flow-based
autoregressive model for molecular graph generation. arXiv preprint arXiv:2001.09382
2020,

(205) Zang, C.; Wang, F. MoFlow: an invertible flow model for generating molecular graphs.
Proceedings of the 26th ACM SIGKDD International Conference on Knowledge Dis-
covery & Data Mining. 2020; pp 617–626.

(206) Luo, Y.; Yan, K.; Ji, S. GraphDF: A Discrete Flow Model for Molecular Graph Gen-
eration. arXiv preprint arXiv:2102.01189 2021,

(207) Vaswani, A.; Shazeer, N.; Parmar, N.; Uszkoreit, J.; Jones, L.; Gomez, A. N.;
Kaiser, L.; Polosukhin, I. Attention is all you need. arXiv preprint arXiv:1706.03762
2017,

(208) Radford, A.; Narasimhan, K.; Salimans, T.; Sutskever, I. Improving language under-
standing by generative pre-training. OpenAI 2018,

(209) Devlin, J.; Chang, M.-W.; Lee, K.; Toutanova, K. Bert: Pre-training of deep bidi-
rectional transformers for language understanding. arXiv preprint arXiv:1810.04805
2018,

60
(210) Radford, A.; Wu, J.; Child, R.; Luan, D.; Amodei, D.; Sutskever, I. Language models
are unsupervised multitask learners. OpenAI blog 2019, 1, 9.

(211) Liu, Y.; Ott, M.; Goyal, N.; Du, J.; Joshi, M.; Chen, D.; Levy, O.; Lewis, M.; Zettle-
moyer, L.; Stoyanov, V. Roberta: A robustly optimized bert pretraining approach.
arXiv preprint arXiv:1907.11692 2019,

(212) Brown, T. B.; Mann, B.; Ryder, N.; Subbiah, M.; Kaplan, J.; Dhariwal, P.; Nee-
lakantan, A.; Shyam, P.; Sastry, G.; Askell, A., et al. Language models are few-shot
learners. arXiv preprint arXiv:2005.14165 2020,

(213) Carion, N.; Massa, F.; Synnaeve, G.; Usunier, N.; Kirillov, A.; Zagoruyko, S. End-to-
end object detection with transformers. European Conference on Computer Vision.
2020; pp 213–229.

(214) Dosovitskiy, A.; Beyer, L.; Kolesnikov, A.; Weissenborn, D.; Zhai, X.; Unterthiner, T.;
Dehghani, M.; Minderer, M.; Heigold, G.; Gelly, S., et al. An image is worth 16x16
words: Transformers for image recognition at scale. arXiv preprint arXiv:2010.11929
2020,

(215) Wang, S.; Guo, Y.; Wang, Y.; Sun, H.; Huang, J. SMILES-BERT: large scale un-
supervised pre-training for molecular property prediction. Proceedings of the 10th
ACM International Conference on Bioinformatics, Computational Biology and Health
Informatics. 2019; pp 429–436.

(216) Honda, S.; Shi, S.; Ueda, H. R. SMILES transformer: pre-trained molecular fingerprint
for low data drug discovery. arXiv preprint arXiv:1911.04738 2019,

(217) Bradshaw, J.; Paige, B.; Kusner, M. J.; Segler, M. H.; Hernández-Lobato, J. M. A
model to search for synthesizable molecules. arXiv preprint arXiv:1906.05221 2019,

61
(218) Grechishnikova, D. Transformer neural network for protein-specific de novo drug gen-
eration as a machine translation problem. Scientific reports 2021, 11, 1–13.

(219) Liu, X.; Zhang, F.; Hou, Z.; Wang, Z.; Mian, L.; Zhang, J.; Tang, J. Self-supervised
learning: Generative or contrastive. arXiv preprint arXiv:2006.08218 2020, 1 .

(220) Wang, Y.; Wang, J.; Cao, Z.; Farimani, A. B. MolCLR: Molecular contrastive learning
of representations via graph neural networks. arXiv preprint arXiv:2102.10056 2021,

(221) Vanschoren, J. Meta-learning: A survey. arXiv preprint arXiv:1810.03548 2018,

(222) Wang, Y.; Yao, Q.; Kwok, J. T.; Ni, L. M. Generalizing from a few examples: A survey
on few-shot learning. ACM Computing Surveys (CSUR) 2020, 53, 1–34.

(223) Kulis, B., et al. Metric learning: A survey. Found. Trends Mach. Learn. 2012, 5,
287–364.

(224) Yang, Z.; Bastan, M.; Zhu, X.; Gray, D.; Samaras, D. Hierarchical Proxy-based Loss
for Deep Metric Learning. arXiv preprint arXiv:2103.13538 2021,

(225) Movshovitz-Attias, Y.; Toshev, A.; Leung, T. K.; Ioffe, S.; Singh, S. No fuss distance
metric learning using proxies. Proceedings of the IEEE International Conference on
Computer Vision. 2017; pp 360–368.

(226) Na, G. S.; Chang, H.; Kim, H. W. Machine-guided representation for accurate graph-
based molecular machine learning. Phys. Chem. Chem. Phys. 2020, 22, 18526–18535.

(227) Koge, D.; Ono, N.; Huang, M.; Altaf-Ul-Amin, M.; Kanaya, S. Embedding of Molec-
ular Structure Using Molecular Hypergraph Variational Autoencoder with Metric
Learning. Mol. Inform. 2020,

(228) Sutton, R. S.; Barto, A. G. Reinforcement learning: An introduction; MIT press, 2018.

62
(229) Arulkumaran, K.; Deisenroth, M. P.; Brundage, M.; Bharath, A. A. A brief survey of
deep reinforcement learning. arXiv preprint arXiv:1708.05866 2017,

(230) Van Hasselt, H.; Guez, A.; Silver, D. Deep reinforcement learning with double q-
learning. Proceedings of the AAAI Conference on Artificial Intelligence. 2016.

(231) Williams, R. J. Simple statistical gradient-following algorithms for connectionist rein-


forcement learning. Machine learning 1992, 8, 229–256.

(232) Schulman, J.; Wolski, F.; Dhariwal, P.; Radford, A.; Klimov, O. Proximal policy
optimization algorithms. arXiv preprint arXiv:1707.06347 2017,

(233) Schulman, J.; Levine, S.; Abbeel, P.; Jordan, M.; Moritz, P. Trust region policy opti-
mization. International Conference on Machine Learning. 2015; pp 1889–1897.

(234) Deng, J.; Yang, Z.; Li, Y.; Samaras, D.; Wang, F. Towards Better Opioid Antagonists
Using Deep Reinforcement Learning. arXiv preprint arXiv:2004.04768 2020,

(235) Yasonik, J. Multiobjective de novo drug design with recurrent neural networks and
nondominated sorting. J. Cheminformatics 2020, 12, 1–9.

(236) Domenico, A.; Nicola, G.; Daniela, T.; Fulvio, C.; Nicola, A.; Orazio, N. De novo drug
design of targeted chemical libraries based on artificial intelligence and pair-based
multiobjective optimization. J. Chem. Inf. Model 2020, 60, 4582–4593.

(237) Liu, X.; Ye, K.; Van Vlijmen, H.; Emmerich, M.; IJzerman, A. P.; van Westen, G.
DrugEx v2: De Novo Design of Drug Molecule by Pareto-based Multi-Objective Re-
inforcement Learning in Polypharmacology. ChemRxiv.14474127.v2 2021,

(238) Reker, D.; Schneider, G. Active-learning strategies in computer-assisted drug discov-


ery. Drug Discov. Today 2015, 20, 458–465.

(239) Walters, W. P.; Murcko, M. Assessing the impact of generative AI on medicinal chem-
istry. Nat. Biotechnol. 2020, 38, 143–145.

63
(240) Sambasivan, N.; Kapania, S.; Highfill, H.; Akrong, D.; Paritosh, P.; Aroyo, L. M.
“Everyone wants to do the model work, not the data work”: Data Cascades in High-
Stakes AI. proceedings of the 2021 CHI Conference on Human Factors in Computing
Systems. 2021; pp 1–15.

(241) Singh, G.; Schulthess, D.; Hughes, N.; Vannieuwenhuyse, B.; Kalra, D. Real world
big data for clinical research and drug development. Drug Discov. Today 2018, 23,
652–660.

(242) Deng, J.; Hou, W.; Dong, X.; Hajagos, J.; Saltz, M.; Saltz, J.; Wang, F. A Large-Scale
Observational Study on the Temporal Trends and Risk Factors of Opioid Overdose:
Real-World Evidence for Better Opioids. Drugs-Real World Outcomes 2021, 1–14.

(243) Deng, J.; Wang, F. An Informatics-based Approach to Identify Key Pharmacological


Components in Drug-Drug Interactions. AMIA Jt Summits Transl Sci Proc 2020,
2020, 142.

(244) Jiang, D.; Wu, Z.; Hsieh, C.-Y.; Chen, G.; Liao, B.; Wang, Z.; Shen, C.; Cao, D.;
Wu, J.; Hou, T. Could graph neural networks learn better molecular representation
for drug discovery? A comparison study of descriptor-based and graph-based models.
J. Cheminformatics 2021, 13, 1–23.

64
Graphical TOC Entry

Some journals require a graphical entry for the


Table of Contents. This should be laid out “print
ready” so that the sizing of the text is correct.
Inside the tocentry environment, the font used
is Helvetica 8 pt, as required by Journal of the
American Chemical Society.
The surrounding frame is 9 cm by 3.5 cm, which
is the maximum permitted for Journal of the
American Chemical Society graphical table of
content entries. The box will not resize if the
content is too big: instead it will overflow the
edge of the box.
This box and the associated title will always be
printed on a separate page at the end of the
document.

65

You might also like