Abstract
Signaling bias is a feature of many G protein-coupled receptor (GPCR) targeting drugs with potential clinical implications. Whether it is therapeutically advantageous for a drug to be G protein biased or β-arrestin biased depends on the context of the signaling pathway. Here, we explored GPCR ligands that exhibit biased signaling to gain insights into scaffolds and pharmacophores that lead to bias. More specifically, we considered BiasDB, a database containing information about GPCR biased ligands, and focused our analysis on ligands which show either a G protein or β-arrestin bias. Five different machine learning models were trained on these ligands using 15 different sets of features. Molecular fragments which were important for training the models were analyzed. Two of these fragments (number of secondary amines and number of aromatic amines) were more prevalent in β-arrestin biased ligands. After training a random forest model on HierS scaffolds, we found five scaffolds, which demonstrated G protein or β-arrestin bias. We also conducted t-SNE clustering, observing correspondence between unsupervised and supervised machine learning methods. To increase the applicability of our work, we developed a web implementation of our models, which can predict bias based on user-provided SMILES, drug names, or PubChem CID. Our web implementation is available at: drugdiscovery.utep.edu/biasnet.
Graphical Abstract
INTRODUCTION
G protein-coupled receptors (GPCRs) represent the largest category of druggable targets for FDA-approved drugs—approximately 34% of all drugs.1,2 GPCRs are signal-transducing proteins that relay information from the extracellular environment of a cell to the cytosol. Upon binding to a drug molecule, GPCRs undergo a conformational change leading an associated G protein to exchange GDP for GTP. Once this exchange occurs, G proteins are activated and they propagate the signaling cascade, ultimately resulting in a cellular response.
G proteins are composed of three subunits: Gα, Gβ, and Gγ. Among these subunits, Gα—the subunit most often responsible for eliciting the cellular/drug response—is categorized into one of four families: Gs, Gi/o, Gq/11, or G12/13.3 Each of these four families activates a specific set of second messengers. For example, Gs stimulates the production of cAMP. In addition to activating G proteins, a GPCR may also activate β-arrestin.4 The preferential activation of either a G protein or β-arrestin is termed biased signaling. In this study, G protein signaling biases present on BiasDB (G protein, G12, Gi, Go, Gq, and Gs) were included in our analysis.
In spite of the extensive druggability of GPCRs, there remains much to be learned about GPCR signaling—both among known and potential GPCR ligands. Signaling bias, a phenomenon first described in 1995, is a feature with the potential of influencing the discovery and approval of novel pharmaceutical agents.5 Because drug molecules targeting a specific signaling pathway minimize the risk of side effects, developing biased drugs leads to safer drugs. In light of this need, many studies have been conducted to characterize cases of GPCR signaling bias.
BiasDB is a manually curated database of such studies.6 BiasDB features 727 cases of biased signaling organized into four bias categories: G protein, G protein selective, extracellular signal-regulated kinases (ERK), and β-arrestin. In addition to compiling known cases of signaling bias, BiasDB also provides a preliminary analysis of the chemical features that tend to make a molecule biased. Knowing these general features can provide a good starting point for drug discovery scientists to develop biased molecules; however, a greater exploration of structure-activity relationship (SAR) is needed. To better describe this SAR, our research objectives were threefold: (1) Build machine learning models. (2) Understand the most important features uncovered by models. (3) Provide examples of clinically relevant drugs that contain scaffolds identified by our models.
First, we used BiasDB to train five machine learning models on 15 different sets of molecular descriptors. We used Random Forest (RF), Multilayer Perception (MLP), XGBoost (XGB), Support Vector Machine (SVM), and Directed Message Passing Neural Network (D-MPNN) algorithms on each set of descriptors. We selected the best-performing set of models that were trained on extended connectivity fingerprints and compared model performance within the five algorithms to determine the best-performing model. Additionally, we tested our top-performing set of models against compounds not found in BiasDB to address the possibility that our models do not generalize to other data sets.
Our team was also interested in determining which, if any, molecular features might be most important to predicting bias. To this end, we trained an MLP model on RDKit features and observed two chemical groups most important for classifying ligands. We noticed that the number of secondary amines and the number of aromatic amines were the most important fragments identified by our model. Knowing this, we compared secondary amines and aromatic amines for G protein biased ligands and β-arrestin biased ligands. We then provided an example of biased and unbiased ligands with differing number of secondary amines to show how these features may explain the SAR of the biased molecules. We corroborated the importance of these features with an unsupervised t-stochastic neighbor embedding (t-SNE) analysis.
We also trained a RF model using chemical scaffolds generated by the HierS method.7 Using these scaffolds as features, we determined which scaffolds are most important for classifying the ligands. Five scaffolds, which show a bias for either β-arrestin or G proteins, are presented here. We conclude with a discussion of these scaffolds as they relate to three biased drugs.
RESULTS AND DISCUSSION
Model Performance.
A total of five different machine learning algorithms along with graph-based neural networks (see Machine Learning section) were trained on the BiasDB data using 15 different descriptors. The best-performing models based on F1 score are reported. The test set scores for each of our five algorithms are shown in Figure 1. For the six metrics used to evaluate our models (F1 score, Recall, Precision, Specificity, Accuracy, and AUC), scores that are closer to 1 indicate better models. Although there is no strict threshold for a “good” model, we can reasonably assume our models were able to correctly classify BiasDB ligands as G protein biased or β-arrestin biased because all metrics for the top-performing models (with the exception of D-MPNN) were above 0.70. All of our models scored an accuracy and AUC above 0.8. Also, all of our models showed a recall above 0.70 and a precision above 0.65. The F1 score for all of our models was above 0.75. The specificity of our models was above 0.8, and for two of our models (XGB and RF), this number was above 0.9. McNemar test results (Table 1) show there is no statistically significant difference between our five models (all pairwise p-values are >0.05). Based on these results, we conclude our models were able to distinguish β-arrestin biased ligands from G protein biased ligands in our training set. Additionally, we observe there was no statistically significant difference between our models’ abilities to distinguish G protein biased ligands from β-arrestin biased ligands.
Table 1.
MLP | XGB | RF | SVM | D-MPNN | |
---|---|---|---|---|---|
MLP | NA | 1 | 1 | 1 | 0.727 |
XGB | 1 | NA | 1 | 1 | 0.774 |
RF | 1 | 1 | NA | 1 | 0.754 |
SVM | 1 | 1 | 1 | NA | 1 |
D-MPNN | 0.727 | 0.774 | 0.754 | 1 | NA |
With these results in mind, it is important to recognize that G protein/β-arrestin bias is a continuum. While the molecules in BiasDB are grouped together in convenient categories that lend themselves to train a classification model (G protein or β-arrestin), it is possible that similar regression models which predict the level of bias can be built. We did not pursue this goal primarily because the metrics used to quantify bias in BiasDB are not consistent from ligand to ligand. For each ligand, two assays are required to determine bias: one that establishes β-arrestin activity and one that establishes G protein activity. While the β-arrestin recruitment assay can be used consistently to determine β-arrestin bias, many different assays can be used to gauge G protein activity (cAMP production, GTPase activity, BRET, and various others). It is because of this reason that we restricted the scope of this study to the classification problem.
External Predictivity.
Having discussed the reliability of our models on BiasDB, we also wanted to demonstrate comparable performance with external test data, thus demonstrating the generalizability of our models. We considered compounds that show potentially therapeutic GPCR signaling bias.3 Out of the compounds discussed in the work done by Tan and colleagues, 21 were not found on BiasDB. The bias of these 21 compounds was predicted using our models. The list of compounds is shown in the Supporting Information. The bias of 18 of these compounds—approximately 90%—was correctly predicted. This finding corroborates the performance of our models on data not found in BiasDB.
Applicability Domain.
Determining whether a compound is biased toward G proteins or β-arrestin is only appropriate when the compound targets GPCRs. To address this concern, the web implementation of our models provides the user information about whether the compound is likely to bind to a GPCR. We developed an SVM machine learning model trained on GPCR-interacting and non-GPCR-interacting ligands to restrict the applicability domain to cases where G protein bias is relevant. Model results are shown in Table 2. Here, we note the applicability domain model used by BiasDB is intended primarily to serve as a warning for the user. The GPCR-binding model is trained on ligands, which bind at the orthosteric sites as well as allosteric sites.
Table 2.
F1 score | recall | precision | specificity | accuracy | AUC |
---|---|---|---|---|---|
0.91 | 0.87 | 0.95 | 0.99 | 0.99 | 0.98 |
Molecular Fragments Related To Bias.
One major goal of our work was to understand intuitively what might make a molecule biased. Molecular fingerprints encode the chemical groups of a molecule into a vector and aid in training accurate models; however, the meaning of this vector is not readily understood by humans. To address this issue, we developed an MLP model trained on RDKit descriptors. The top-20 most important RDKit descriptors were analyzed, and from this list, we observed the two most important fragments are the number of secondary amines and the number of aromatic amines.
Descriptive statistics summarizing the number of secondary amines and aromatic amines for BiasNet ligands are shown in Table 3. The table shows ligands in BiasDB had 0 to 14 secondary amines. While this first range is large, it is worth noting that 75% of G protein biased ligands had zero or one secondary amine and 75% of β-arrestin biased ligands had zero, one, or two secondary amines. The range of aromatic amines is markedly smaller, however. This range is 0–3 aromatic amines. We observe aromatic amines are also more common in β-arrestin biased ligands. In particular, the value at Quartile 3 (Q3) for G protein biased aromatic amines is 0, and the value at Q3 for β-arrestin aromatic amines is 1.
Table 3.
bias | mean | min | Q1 | median | Q3 | max | |
---|---|---|---|---|---|---|---|
secondary amines | G protein | 1.07 | 0 | 0 | 1 | 1 | 14 |
β-arrestin | 2.44 | 0 | 1 | 1 | 2 | 12 | |
aromatic amines | G protein | 0.22 | 0 | 0 | 0 | 0 | 2 |
β-arrestin | 0.49 | 0 | 0 | 0 | 1 | 3 |
The mean number of secondary amines for G protein biased ligands was 1.07, while for β-arrestin biased ligands, it was 2.44. The mean for G protein aromatic amines was 0.22, and for β-arrestin, this number was 0.49. One-tailed Z-tests comparing the difference in means for secondary amines and for aromatic amines show a statistically significant difference between the averages of β-arrestin biased ligands and G protein biased ligands (p-value < 0.01 for both of these fragments).
Figure 2 more completely shows the distribution of molecular fragments. The violin plot on the left of Figure 2a exhibits one high-density region at the base, corresponding to 0–1 secondary amines. The violin plot on the right has two such regions. The first region is at the base of the plot and has the greatest width at around 1 secondary amine. The second region is at the top of the plot and has the greatest width at 10 secondary amines. Figure 2b shows that G protein biased ligands had the greatest density at 0 for aromatic amines. Similarly, β-arrestin biased ligands had the greatest density at 0 aromatic amines. The difference is that for β-arrestin biased ligands, a greater proportion of compounds have 1 or 2 aromatic amines.
Taken together, these statistics suggest increased secondary amines and aromatic amines might be connected to β-arrestin bias relative to G protein bias. While our models did identify this pattern, we recognize that other features play a larger role in determining bias. Thus, it is not possible to say with complete certainty that a molecule with no secondary amines or no aromatic amines is G protein biased. Similarly, it is not possible to say that a molecule with many secondary amines or aromatic amines is β-arrestin biased—merely that these fragments tend to be found in greater abundance in β-arrestin biased ligands.
In addition to highlighting these statistics, our aim was to try to understand the structural reasons that might lead to bias. We focus our discussion on secondary amines and consider the two molecules in Figure 3. Both molecules are ethanolamines and agonists for the β-2 adrenergic receptor (β2R), and have a similar mechanism of action. The molecule on the top panels (isoproterenol) contains only one secondary amine, while the molecule on the bottom panels (BI-167107) contains two secondary amines. The molecule on the top panels is unbiased, while the molecule on the bottom panels is β-arrestin biased.
Previous docking studies have demonstrated that isoproterenol can form hydrogen bonds between its para hydroxyl and SER 203, SER 204, and SER 207 on Transmembrane 5 (TM5) of the β-2 adrenergic receptor.8 Additionally, the meta-hydroxyl group can form hydrogen bonds with SER 203. We suspect weak interactions with TM5 might be connected to β-arrestin bias.
We performed docking studies on isoproterenol and compared them to BI-16707 co-crystallized to the β-2 adrenergic receptor. Out of all docking poses, we chose the one with the second-lowest binding energy as it closely resembles the pose reported in the literature by Weiss and colleagues.8 All binding poses and associated binding energies are available in the Supporting Information Figure S1. The binding poses of the two compounds are shown in Figure 3. Like in the study by Weiss et al., the para-hydroxyl group on isoproterenol appears in close proximity to SER 203 and SER 204; however, our model does not find a hydrogen bond between the para-hydroxyl group and SER 204. Still, our simulation did find a hydrogen bond between the meta-hydroxyl and SER 203. Importantly, in both the docking simulations done by Weiss et al.8 and in this study, isoproterenol explicitly interacts with TM5.
Co-crystallized BI-16707 shows similar proximity to SER 203 and SER 204 with its para-hydroxyl group and a secondary ring member amine; however, the co-crystallized structure did not demonstrate any hydrogen bond. While it is still possible that BI-16707 interacts with TM5 residues via dipole–dipole interactions, these are not as strong as hydrogen bonds.
Another factor to consider is the role the ring member nitrogen plays in determining bias. Whether this nitrogen participates in dipole–dipole interactions or hydrogen bonds with residues from TM5, such interactions would be weaker than analogous interactions with a hydroxyl from the isoproterenol group due to the higher electronegativity of oxygen.
We believe the stronger interaction of isoproterenol with TM5 might account for the balanced bias demonstrated by isoproterenol. By the same token, the weaker interactions between BI-167107 might explain β-arrestin bias. Experiments showing isoproterenol co-crystallized with the β-2 adrenergic receptor are needed to clarify the role of SER 203, SER 204, and SER 207 and confirm our hypothesis about β-arrestin bias.
SCAFFOLDS CONTRIBUTING TO BIAS
In addition to considering molecular features, our goal was also to utilize a machine learning model trained on scaffolds. The top-five scaffolds identified by our model are shown in Figure 4. We note that scaffolds 1 and 2 have no secondary amines (scaffold 2 has substituents replacing the amine hydrogen for all ligands), and scaffolds 3, 4, and 5 have 1 secondary amine (scaffold 3 has a methyl substituent replacing the nitrogen on the six-membered ring). These numbers are consistent with the previous findings about molecular fragments: scaffolds 1 and 2 are G protein biased and have no secondary amines, and scaffolds 3, 4, and 5 have one secondary amine.
Scaffold 1 is the molecule pyridine. There are 123 cases of G protein or β-arrestin bias on BiasDB with the pyridine substructure. Of these cases, 79 are G protein biased and only 13 are β-arrestin bias. Scaffold 1 is overwhelmingly present in cases of G protein bias. Scaffold 1 is present in ligands that bind to 10 different receptors (5-HT1, 5-HT2, CB2, CXCR3, D1, D2, Ghrelin, LH, Kappa, and Mu receptor).
Scaffold 2, 1-(2-pyridinyl)piperazine, is a super-scaffold containing pyridine (scaffold 1). There are 17 cases of bias on BiasDB with the scaffold 2 substructure. Of these cases, 1 is an ERK bias and 16 are G protein biases. Scaffold 2 was found in ligands that bind D2 and 5-HT1 receptors. Importantly, all ligands on BiasDB containing Scaffold 2 have a substituent replacing the amine hydrogen. Thus, scaffold 2 does not contribute any secondary amines to ligands.
Scaffold 3 is an ergot alkaloid-like molecule. There are 8 cases of bias on BiasDB with scaffold 3. Of these, 7 cases are associated with β-arrestin bias and 1 case is associated with G protein selectivity (that is, a bias toward one particular G protein subtype over another subtype). Scaffold 3 is present in molecules that bind to the 5HT2B and D2 receptors. One noteworthy remark about scaffold 3 is that mechanistic details about bias at the 5HT2B receptor have previously been characterized.9 Researchers have found evidence that amine residues might be important in imparting β-arrestin bias to certain classes of molecules. In particular, the secondary amine in the six-membered ring forms a hydrogen bond to the amino acid backbone of GLY 221 (G5.42). These interactions, together with the bulky size of the scaffold, allow molecules like ergotamine to shape the confirmation of the extracellular loop (EL) region such that they lead to β-arrestin bias.
Scaffold 4, 4,5-dihydro-1H-imidazole, is a five-member two-nitrogen heterocycle with one double bond. There are 8 cases of bias on BiasDB with scaffold 4. Of these, 4 cases are associated with β-arrestin bias and the remaining cases are associated with G protein selectivity or ERK bias. There are no cases of G protein bias relative to β-arrestin bias, so we have reason to believe the scaffold is associated with β-arrestin bias. Scaffold 4 is present in molecules that bind to the α1A and α2C adrenoceptors.
Scaffold 5 is a right-hand side bicyclic aromatic moiety found in many of the compounds.10 There are 5 cases of bias on BiasDB with this structure. Of these cases, 4 are β-arrestin bias and only one exhibits G protein bias. In the work done by Chen et al.,10 variations of the bicyclic aromatic moiety still led to β-arrestin bias to different degrees.
Unsupervised Learning.
To corroborate the findings of our supervised machine learning models, we performed an unsupervised machine learning technique, t-stochastic neighbor embedding (t-SNE). We consider the 5 clusters in our discussion represented in Figure 5. Templates for each of the five clusters are shown in Figure 6. A full collection of cluster compounds can be found in the Supporting Information.
Cluster 1 (enclosed by a red box in Figure 5) identifies 15 G protein selective EP2 agonists. These ligands (represented in Figure 6) share three features that form a common structural backbone. The SAR of these compounds has previously been described by researchers synthesizing novel EP2 selective agonists.11 The first feature is a thiazole ring, which is important for EP2 receptor subtype selectivity.12 The second feature is a middle bicyclic moiety, which allows the molecules to mimic endogenous prostacyclins. The third feature is a variable ω chain (corresponding to the R1 group), which contributes to bias. Importantly, we note that these ligands do not contain secondary amines consistent with the idea that G protein biased ligands tend to have less of these moieties than β-arrestin biased compounds.
Cluster 2 identifies G protein biased κ opioid receptor agonists. These ligands share a common central triazole which forms a scaffold attached to three different aromatic rings. One of these rings is a 4-pyridyl ring, and the remaining two are variable aromatic rings. The SAR of these compounds has previously been described by researchers investigating biased κ opioid receptor agonists.13,14 To alter bias, the non-pyridyl rings are exchanged, but the core triazole ring is left intact. The core triazole also contains no secondary amines.
Cluster 3 represents G protein biased D1 receptor agonists. Of the 12 ligands in this cluster, all are G protein biased. Researchers have previously described the binding mode of these compounds, referred to in the literature as non-catechol D1 agonists.15 These ligands interact with SER 188 and LEU 190 from intracellular loop 2 (ECL2) via the variable R3 group.
Cluster 4 represents β-arrestin biased D2 receptor agonists. Cluster 4 contains 17 ligands, 16 of which are β-arrestin biased. Aripiprazole is the exception to this cluster and is the unbiased reference compound by which biased compounds are compared. The SAR of these compounds has previously been characterized.10,16–18 Each of these ligands can be divided into four regions: a “left-hand side (LHS) phenyl ring,” “middle cyclic amino moiety,” “central linker,” and “right-hand side (RHS) bicyclic aromatic moiety.”10 The region we will discuss is the RHS bicyclic aromatic moiety pictured in Figure 6. This moiety contains one secondary amine. Interestingly, the work done by Chen and colleagues found one compound (not included in this cluster) for which the RHS bicyclic aromatic moiety is replaced with a benzimidazolone group. This compound, 5-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-butoxy)-1H-benzo[d]imidazol-2(3H)-one, has two secondary amines and shows an increased β-arrestin bias relative to aripiprazole, further highlighting the connection between secondary amines and β-arrestin bias.
Cluster 5 contains 19 ligands, 18 of which are G protein biased. Among the receptors targeted by cluster 5 are the μ-opioid, κ-opioid, nociceptin, apelin, and ghrelin receptors. We limit our discussion to μ-opioid receptors agonists, as the majority (13) of ligands from cluster 5 are μ-opioid agonists. The μ-opioid ligands in cluster 5 are G protein biased and have the common structure, as shown in Figure 6. All variations of these compounds have at most 1 secondary amine—the amine in the 5-member ring. While this finding does not definitively require that all G protein biased μ-opioid receptor targeting compounds have at most 1 secondary amine, it further highlights the connection we observed between a small number of secondary amines and G protein bias.
Clinically Significant Compounds with HierS Scaffolds.
To end our discussion of scaffolds, we chose to illustrate how bias can have a clinically significant effect. Here, we consider three commercially available medications, as shown in Figure 7. Oliceridine is a recently FDA-approved G protein biased drug molecule containing scaffold 1. Oliceridine is a μ-opioid receptor agonist intended to treat severe acute pain. β-Arrestin activity for μ-opioid receptors is associated with increased gastrointestinal dysfunction, respiratory dysfunction, and decreased efficacy.19 Because oliceridine shows a marked G protein bias, researchers suspect oliceridine might have a better safety profile than commercially available drugs like morphine. Despite this optimism, other researchers suggest the enhanced safety profile of oliceridine might be attributed to a lower intrinsic activity for the drug.20 In any case, more research is needed to confirm whether oliceridine is a suitable alternative for current opioid drugs.
Lisuride is a β-arrestin biased drug containing scaffold 3. Lisuride binds to D2 receptors and is indicated for Parkinson’s disease. Standard treatments for Parkinson’s include the drug l-Dopa, which activates dopamine receptors in the body. l-Dopa is associated with side effects such as dyskenesia, which are thought to be caused by receptor supersensitivity.21 Lisuride bypasses such side effects because its β-arrestin action mitigates this receptor supersensitivity. For this reason, lisuride is often prescribed before l-Dopa.
Clonidine is a β-arrestin biased drug containing scaffold 4. Clonidine binds to the α2 adrenergic receptor and is indicated for high blood pressure. Work has been conducted for related groups of anti-hypertensive and cardio-protective drugs targeting the β1 adrenergic receptors. This work links β-arrestin activation to “therapeutically advantageous” activity.22 Although clonidine works through a different receptor and overall mechanism of action, there is reason to believe β-arrestin bias for clonidine could also be a desirable characteristic for the drug. For clonidine, β-arrestin bias is liked to increase endocytosis of the α2 adrenergic receptor.23 This β-arrestin-directed effect presumably leads to desensitization of the ligand, which results in decreased drug side effects.
CONCLUSIONS
In summary, our work was geared toward determining if there are global patterns of bias among ligands present in the recently available database BiasDB. BiasDB contains cases of bias for many different receptor types and many different ligand types with different SARs. Our machine learning models accurately picked up on these global patterns and correctly categorized ligands as G protein biased ligands or β-arrestin biased ligands; therefore, we had reason to believe there likely exist chemical groups which are found in greater quantities in either case of β-arrestin or G protein bias.
We determined from our models that secondary amines are chemical features, which are more likely to be found in β-arrestin biased compounds compared to G protein biased compounds. Also, we trained a separate model on HierS scaffolds and found that 2 scaffolds were related to G protein bias and 3 were related to β-arrestin bias. The two β-arrestin biased scaffolds had secondary amines. Lastly, we utilized t-SNE clustering and found that within four G protein clusters, the majority of compounds had either zero or one secondary amine. In one β-arrestin cluster, all compounds had secondary amines.
With these findings in mind, we hope that our work can be used by drug discovery scientists to develop more efficacious drug therapies with less side effects. Web implementation of our model is freely available at: drugdiscovery.utep.edu/biasnet.
METHODS
Molecular Features.
Supervised machine learning models included those trained solely on molecular descriptors and one trained solely on scaffolds. In total, 15 molecular descriptors (each represented by a vector of different size) were used to train each of 15 descriptor models, and one vector with 704 scaffolds was used to train a scaffold model. The model used to make predictions on the Biasnet web service uses the long ECFP4 vector described below. The scaffolds used to train the scaffolds model are discussed in the following Scaffolds section. The unsupervised t-SNE model was trained solely on RDK7 fingerprints, as the t-SNE algorithm relies on dimensionality reduction, and using multiple descriptors would make this task more difficult. Here, we discuss each of the descriptors in more detail.
Extended Connectivity Fingerprints.
Extended connectivity fingerprints (ECFPs) are a class of circular fingerprints, which are generated by considering a diameter of a given size around each atom in a molecule.24 The greater the diameter, the larger the chemical environment about each individual atom. The chemical environment can be encoded into a vector using a ECFP-specific hash function. Our project used ECFP2, ECFP4, and ECFP6 from RDKit, which correspond to diameters 2, 4, and 6, respectively. The vector sizes used were 1024 bit (short) and 16 384 bit (long). Our project used ECFP2 (short), ECFP4 (long), and ECFP6 (long and short), totaling 4 ECFP vectors.
Functional Class Fingerprints.
Functional class fingerprints (FCFP) are very similar to ECFPs, the difference being that FCFPs take into account the chemical roles of atoms.24 For instance, a nitrogen atom might be encoded as a hydrogen bond-acceptor or a hydrogen bond-donor, depending on the chemical context of the nitrogen atom. Our project used FCFP4 and FCFP6 from RDKit, which correspond to diameters of 4 and 6, respectively. The size of both fingerprints was 16384 bits.
Morgan Fingerprints.
Morgan fingerprints, also like ECFPs, are a class of circular fingerprint. The Morgan Algorithm is used to construct the vector representation for the fingerprints.25 Our team used Morgan fingerprints from RDKit with radius 2 and vector length 1024.
Avalon Fingerprints.
Avalon fingerprints are a class of substructure key fingerprint.26 A substructure key fingerprint is a vector of fixed size, where each position in the vector corresponds to a chemical substructure. The Avalon fingerprint vectors used were from RDKit, and were of length of 1024 (short) and 16 384 (long).
Molecular Access System Keys.
Like Avalon fingerprints, molecular access system keys (MACCS Keys) are a kind of substructure key. Instead of a vector size of 512, MACCS keys are of size 167. Our team used MACCS Keys from RDKit.27
RDK5 Fingerprints.
RDK5 Fingerprints are a class of path-based fingerprints provided by RDKit. Path-based fingerprints are generated by following a path of maximum bond length from a starting atom. All of the fragments generated are encoded to a vector of predefined size. Our team used a maximum bond length of 5 and a vector length of 2048.
Topological Pharmacophore Atomic Pair Fingerprints.
Topological pharmacophore atomic triplet fingerprints (TPAPFs) are a kind of path-based fingerprint.28–30 The algorithm used to encode the fingerprints relies on finding pairs of descriptor centers of a molecule (usually heteroatoms, unsaturated bonds, or other pharmacophores) separated by a maximum bond length. These are then encoded into a vector of a fixed size. Our team used the TPAPFs obtained with MayaChemTools. The vector size was 150.
Topological Pharmacophore Atomic Triplet Fingerprints.
Topological pharmacophore atomic triplet fingerprints (TPATFs) are a kind of path-based fingerprint similar to TPAPFs, but instead of using pharmacophore pairs, they use triplets.31 TPATFs were also obtained using MayaChemTools. The vector size was 2692.
RDkit Descriptors.
RDKit descriptor is a list of one-dimensional molecular features such as MolecularWeight, HeavyAtoms, MolecularVolume, RotatableBonds, Hydrogen-BondDonor, HydrogenBondAcceptors, S Log P, and topological polar surface area (TPSA). In total, 200 descriptors were used.
MayaChemTools Physicochemical Properties.
Mayachem-tools physicochemical properties is a list of one-dimensional molecular features. In total, 8 descriptors were used. The eight features used include ring count, rotatable bonds,and fraction of SP3 carbons.
Scaffolds.
All queried G protein biased ligands and β-arrestin biased ligands were introduced into the hscaf web app available at http://pasilla.health.unm.edu/tomcat/hscaf/hscaf. After processing, 704 scaffolds were generated. Thus, a 704-bit vector was used to train a RF model, where the presence of a scaffold was denoted by a 1 in the vector, and the absence was denoted by a 0. The scaffolds are included in the Supporting Information.
Following model development, importance scores were calculated using the permutation feature importance module found in Scikit-learn. Scaffolds with a score greater than 0.005 were chosen for screening. Selection based on importance score resulted in 10 scaffolds. Out of these, scaffolds were cross-checked against BiasDB to ensure that the scaffolds represented primarily one type of bias. Five scaffolds fit this criterion.
Machine Learning.
The main data set was divided into training (80%) and test (20%) sets using a stratified sampling method, which ensures the equal distribution of different classes into both sets. Firstly, four different machine learning (ML) algorithms, namely random forest (RF), extreme gradient boosting, multilayer perceptron (MLP), and support vector machine (SVM) were trained with their default settings, with 15 different descriptors (see Molecular Descriptors). Finally, the best-suited combination of classifiers and features (RF/Avalon, XGB/Avalon, MLP/lecfp4, and SVM/ecfp2) were selected and further tuned based on hyperparameter optimization. We used Hypeopt,32 where each model was tuned based on the grid search method, with default 3-fold cross-validation. To compare the model performances between the models trained with manually computed molecular fingerprints and models trained on graph-based neural networks that learn by operating on the graph structure of the molecule, we retrained the models using Chemprop D-MPNN. The D-MPNN model was also trained with 3-fold cross-validation. It was optimized using various model hyperparameters such as the hidden size of the neural network layers, models depth, number of feedforward network layers, and dropout probability. Among all of the optimized models, the outperforming model based on the test set F1 score was used for web implementation. The five aforementioned ML algorithms are summarized as follows.
Random Forest.
Random forest is a supervised learning algorithm used for both classification and regression problems. It comprises multiple decision trees with the technique called bootstrap aggregation (also known as bagging) and gets a prediction from each tree outputting the best solution by means of voting. The model was tuned with mainly three hyperparameters (n_estimators, min_samples_split, and max_features) to get the optimized model.
XGBoost.
It is also a decision tree-based ensemble algorithm that uses a gradient boosting framework designed for speed and performance. The training of the model can be parallelized/distributed across the clusters. The output of the model is obtained by summing up a sequence of decision trees, called boosting. Each new tree corrects errors which were made by the previously trained decision tree. The best model was achieved by tuning three different hyperparameters (n_estimators, learning_rate, and colsample_bytree).
Multilayer Perceptron.
Multilayer perceptron (MLP) is a feedforward artificial neural network (ANN) capable of mapping the input data into appropriate outputs. ANNs are brain-inspired systems comprised of artificial neurons commonly known as nodes. A MLP consists of an input layer, hidden layers, and output layer, and each layer is fully connected to the following layer. Each neuron is applied to an activation function except for the input layer. We used a rectified linear unit (ReLU) as an activation function and “adam” as a solver or weight optimization.
Support Vector Machine (SVM).
By extending the feature space using quadratic, cubic, and higher-order polynomial functions of the predictors, the support vector machine (SVM) method addresses a nonlinear boundary between groups. By finding the hyperplane, which maximizes the margin, SVM handles the classification. The margin is the difference in the direction common to the hyperplane between the two nearest instances from the opposite groups. SVM can scan through the maximum margin between two groups for this boundary line, and the distance between the line and both of these points is equidistant. Therefore, in order for the line to be the product of the SVM, the sum of these two distances needs to be maximized.
Direct Message Passing Neural Network (D-MPNN).
The direct message passing neural network (D-MPNN)33 is a graph-based neural network built on the framework of MPNN by Gilmer et al.34 An MPNN is a unified framework to generalize several graph convolution neural network methods. The input to MPNN is an undirected graph G with node features xv and bond features evw, where v and w are the node indices. The framework consists of two phases: the message passing phase and the readout phase. The message passing phase gathers the structural information of the graph and the readout phase uses that information of the graph to make predictions. The D–MPNN differs from MPNN in terms of messages passing through the network. D-MPNN updates the information of the directed bonds, whereas MPNN updates the information of atoms.
Machine Learning Model Comparison.
Our Machine learning models were evaluated by receiver operation characteristic area under the curve (ROC AUC), accuracy, F1 score, recall, precision, and specificity. Additionally, a pairwise McNemar test was conducted to compare the performance of each machine model against all of the other models. These metrics are calculated with true positive (TP), true negative (TN), false positive (FP), and false negative (FN) classifications.
Accuracy.
Accuracy is the ratio of correctly predicted items divided by the total number of items.
Recall.
Recall is the ratio of true positives over total positives.
Precision.
Precision is the ratio of true positives over predicted positives.
Specificity.
Specificity is the ratio of true negatives over total negatives.
F1 Score.
Specificity is the harmonic mean of precision and recall.
ROC AUC.
ROC AUC can be calculated by integrating the curve of the true positive rate as a function of the false positive rate. For more details, the reader can consult other works, which use the ROC AUC metric.35
Applicability Domain.
The target central resource database version 540 (TCRD v540) was used for training the applicability domain model. The compound activity table was merged with the protein table to obtain a mapping of ligand to target. For this combined table, rows with target descriptions containing the phrase “G protein-coupled” were assigned a label of 1. Rows not containing the previous description were given the label 0. The data set was featurized using long ECFP4s. A linear SVM model was trained and tested on these features following a 75/25 train/test split. The regularization hyperparameter C was set to 1 during model development. In total, 381 652 ligands were used, 709 of which showed activity for GPCRs and 380 943 for non-GPCRs.
Supplementary Material
ACKNOWLEDGMENTS
The authors thank Jeremy Yang from the Translational Informatics Division at the University of New Mexico (UNM) School of Medicine for helping their team implement hscaf and Hassan Mahmudulla for providing support for machine learning model development and web hosting.
Funding
Dr. Sirimulla acknowledges support from the National Science Foundation through NSF-PREM Grant DMR-1827745. G.B.K., J.S, and J.G are supported by NSF-PREM Grant DMR-1827745. J.F. is supported through UTEP BUILDing Scholars program funded by NIH under linked Award Numbers RL5GM118969, TL4GM118971.
Footnotes
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jcim.1c00317.
Additional binding pose information and chemical structures for cluster compounds (PDF)
Scaffolds and compounds used for external predictivity (XLSX)
Complete contact information is available at: https://pubs.acs.org/10.1021/acs.jcim.1c00317
The authors declare no competing financial interest.
A web-based application can be accessed at https://drug/discovery.utep.edu/biasnet/. All of the code and data are available at: https://github.com/sirimullalab/biasNet. Third-party software used for model development can be found under the environment.yml file under the same GitHub page. All molecular docking experiments were performed using AutoDock Vina version 1.1.2.36 The β-2 Adrenergic Receptors (entries 6KR8 and 3P0G) were both obtained from the Protein Data Bank. Two-dimensional (2D) visualization was accomplished with Maestro. Three-dimensional (3D) visualization was accomplished with PyMOL. A prototype version of BiasNet is in development for the University of Texas Research Cyberinfrastructure (UTRC) web portal: https://utrc.tacc.utexas.edu/. The in-development version supports large-scale and high-throughput ligand evaluation, backed by clusters at the Texas Advanced Computer Center.
Contributor Information
Julian Franco, Mechanical Engineering, The University of Texas at El Paso, El Paso, Texas 79968, United States.
William J. Allen, Texas Advanced Computing Center, The University of Texas at Austin, Austin, Texas 78758, United States
Jesus David Garcia, Computer Science, The University of Texas at El Paso, El Paso, Texas 79968, United States.
Suman Sirimulla, Department of Pharmaceutical Science and Computational Science Program, The University of Texas at El Paso, El Paso, Texas 79968, United States; Computer Science, The University of Texas at El Paso, El Paso, Texas 79968, United States.
REFERENCES
- (1).Sriram K; Insel PA G protein-coupled receptors as targets for approved drugs: How many targets and how many drugs? Mol. Pharmacol 2018, 93, 251–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (2).Hauser AS; Attwood MM; Rask-Andersen M; Schiöth HB; Gloriam DE Trends in GPCR drug discovery: New agents, targets and indications. Nat. Rev. Drug Discovery 2017, 16, 829–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (3).Tan L; Yan W; McCorvy JD; Cheng J Biased Ligands of G Protein-Coupled Receptors (GPCRs): Structure-Functional Selectivity Relationships (SFSRs) and Therapeutic Potential. J. Med. Chem 2018, 61, 9841–9878. [DOI] [PubMed] [Google Scholar]
- (4).Reiter E; Ahn S; Shukla AK; Lefkowitz RJ Molecular Mechanism of β-Arrestin-Biased Agonism at Seven-Transmembrane Receptors. Annu. Rev. Pharmacol. Toxicol 2012, 52, 179–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (5).Kenakin T Agonist-receptor efficacy II: agonist trafficking of receptor signals. Trends Pharmacol. Sci 1995, 16, 232–238. [DOI] [PubMed] [Google Scholar]
- (6).Omieczynski C; Nguyen N; Sribar D; Deng L; Stepanov D; Schaller D; Wolber G; Bermudez M BiasDB: AComprehensive Database for Biased GPCR Ligands. bioRxiv 2019, No. 742643. [Google Scholar]
- (7).Wilkens SJ; Janes J; Su AI HierS: Hierarchical scaffold clustering using topological chemical graphs. J. Med. Chem 2005, 48, 3182–3193. [DOI] [PubMed] [Google Scholar]
- (8).Weiss DR; Ahn S; Sassano MF; Kleist A; Zhu X; Strachan R; Roth BL; Lefkowitz RJ; Shoichet BK Conformation guides molecular efficacy in docking screens of activated β-2 adrenergic G protein coupled receptor. ACS Chem. Biol 2013, 8, 1018–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (9).Denzinger K; Nguyen TN; Noonan T; Wolber G; Bermudez M Biased ligands differentially shape the conformation of the extracellular loop region in 5-HT2B receptors. Int. J. Mol. Sci 2020, 21, No. 9728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (10).Chen X; Sassano MF; Zheng L; Setola V; Chen M; Bai X; Frye SV; Wetsel WC; Roth BL; Jin J Structure-functional selectivity relationship studies of β-arrestin-biased dopamine D2 receptor agonists. J. Med. Chem 2012, 55, 7141–7153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (11).Ogawa S; Watanabe T; Sugimoto I; Moriyuki K; Goto Y; Yamane S; Watanabe A; Tsuboi K; Kinoshita A; Kigoshi H; Tani K; Maruyama T Discovery of G Protein-Biased EP2 Receptor Agonists. ACS Med. Chem. Lett 2016, 7, 306–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (12).Kambe T; Maruyama T; Nakai Y; Yoshida H; Oida H; Maruyama T; Abe N; Nishiura A; Nakai H; Toda M Discovery of novel prostaglandin analogs as potent and selective EP2/EP4 dual agonists. Bioorg. Med. Chem 2012, 20, 2235–2251. [DOI] [PubMed] [Google Scholar]
- (13).Lovell KM; Frankowski KJ; Stahl EL; Slauson SR; Yoo E; Prisinzano TE; Aubé J; Bohn LM Structure–Activity Relationship Studies of Functionally Selective Kappa Opioid Receptor Agonists that Modulate ERK 1/2 Phosphorylation While Preserving G Protein Over βArrestin2 Signaling Bias. ACS Chem. Neurosci 2015, 6, 1411–1419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (14).Zhou L; Lovell KM; Frankowski KJ; Slauson SR; Phillips AM; Streicher JM; Stahl E; Schmid CL; Hodde P; Madoux F; Cameron MD; Prisinzano TE; Aubé J; Bohn LM Development of functionally selective, small molecule agonists at kappa opioid receptors. J. Biol. Chem 2013, 288, 36703–36716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (15).Gray DL; Allen JA; Mente S; O’connor RE; Demarco GJ; Efremov I; Tierney P; Volfson D; Davoren J; Guilmette E; Salafia M; Kozak R; Ehlers MD Impaired β-arrestin recruitment and reduced desensitization by non-catechol agonists of the D1 dopamine receptor. Nat. Commun 2018, 9, No. 674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (16).Vangveravong S; Zhang Z; Taylor M; Bearden M; Xu J; Cui J; Wang W; Luedtke RR; MacH RH Synthesis and characterization of selective dopamine D2 receptor ligands using aripiprazole as the lead compound. Bioorg. Med. Chem 2011, 19, 3502–3511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (17).Oshiro Y; Sato S; Kurahashi N; Tanaka T; Kikuchi T; Tottori K; Uwahodo Y; Nishi T Novel antipsychotic agents with dopamine autoreceptor agonist properties: Synthesis and pharmacology of 7-[4-(4-phenyl-1- piperazinyl)butoxy]-3,4-dihydro-2(1H)-quinolinone derivatives. J. Med. Chem 1998, 41, 658–667. [DOI] [PubMed] [Google Scholar]
- (18).Johnson DS; Choi C; Fay LK; Favor DA; Repine JT; White AD; Akunne HC; Fitzgerald L; Nicholls K; Snyder BJ; Whetzel SZ; Zhang L; Serpa KA Discovery of PF-00217830: Aryl piperazine napthyridinones as D2 partial agonists for schizophrenia and bipolar disorder. Bioorg. Med. Chem. Lett 2011, 2621–2625. [DOI] [PubMed] [Google Scholar]
- (19).DeWire SM; Yamashita DS; Rominger DH; Liu G; Cowan CL; Graczyk TM; Chen XT; Pitis PM; Gotchev D; Yuan C; Koblish M; Lark MW; Violin JD A G protein-biased ligand at the μ-opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with morphines. J. Pharmacol. Exp. Ther 2013, 344, 708–717. [DOI] [PubMed] [Google Scholar]
- (20).Gillis A; Gondin AB; Kliewer A; Sanchez J; Lim HD; Alamein C; Manandhar P; Santiago M; Fritzwanker S; Schmiedel F; Katte TA; Reekie T; Grimsey NL; Kassiou M; Kellam B; Krasel C; Halls ML; Connor M; Lane JR; Schulz S; Christie MJ; Canals M Low intrinsic efficacy for G protein activation can explain the improved side effect profiles of new opioid agonists. Sci. Signaling 2020, 13, No. eaaz3140. [DOI] [PubMed] [Google Scholar]
- (21).Urs NM; Bido S; Peterson SM; Daigle TL; Bass CE; Gainetdinov RR; Bezard E; Caron MG Targeting β-arrestin2 in the treatment of L-DOPA-induced dyskinesia in Parkinson’s disease. Proc. Natl. Acad. Sci. USA 2015, 112, E2517–E2526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (22).Carr R; Schilling J; Song J; Carter RL; Du Y; Yoo SM; Traynham CJ; Koch WJ; Cheung JY; Tilley DG; Benovic JL β-arrestin-biased signaling through the β2-adrenergic receptor promotes cardiomyocyte contraction. Proc. Natl. Acad. Sci. USA 2016, 113, E4107–E4116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (23).Lu R; Li Y; Zhang Y; Chen Y; Shields AD; Winder DG; Angelotti T; Jiao K; Limbird LE; Zhou Y; Wang Q Epitope-tagged receptor knock-in mice reveal that differential desensitization of α2-adrenergic responses is because of ligand-selective internalization. J. Biol. Chem 2009, 284, 13233–13243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (24).Rogers D; Hahn M Extended-connectivity fingerprints. J. Chem. Inf. Model 2010, 50, 742–754. [DOI] [PubMed] [Google Scholar]
- (25).Morgan HL The Generation of a Unique Machine Description for Chemical Structures–A Technique Developed at Chemical Abstracts Service. J. Chem. Doc 1965, 5, 107–113. [Google Scholar]
- (26).Gedeck P; Rohde B; Bartels C QSAR - How good is it in practice? Comparison of descriptor sets on an unbiased cross section of corporate data sets. J. Chem. Inf. Model 2006, 46, 1924–1936. [DOI] [PubMed] [Google Scholar]
- (27).Durant JL; Leland BA; Henry DR; Nourse JG Reoptimization of MDL keys for use in drug discovery. J. Chem. Inf. Comput. Sci 2002, 42, 1273–1280. [DOI] [PubMed] [Google Scholar]
- (28).Schneider G; Neidhart W; Giller T; Schmid G “Scaffold-Hopping” by Topological Pharmacophore Search: A Contribution to Virtual Screening. Angew. Chem., Int. Ed 1999, 38, 2894–2896. [PubMed] [Google Scholar]
- (29).Fechner U; Franke L; Renner S; Schneider P; Schneider G Comparison of correlation vector methods for ligand-based similarity searching. J. Comput.-Aided Mol. Des 2003, 17, 687–698. [DOI] [PubMed] [Google Scholar]
- (30).Fechner U; Schneider G Evaluation of Distance Metrics for Ligand-Based Similarity Searching. ChemBioChem 2004, 5, 538–540. [DOI] [PubMed] [Google Scholar]
- (31).McGregor MJ; Muskal SM Pharmacophore fingerprinting. 1. Application to QSAR and focused library design. J. Chem. Inf. Comput. Sci 1999, 39, 569–574. [DOI] [PubMed] [Google Scholar]
- (32).Hypopt. https://github.com/cgnorthcutt/hypopt (accessed May 17th, 2021).
- (33).Yang K; Swanson K; Jin W; Coley C; Eiden P; Gao H; Guzman-Perez A; Hopper T; Kelley B; Mathea M; Palmer A; Settels V; Jaakkola T; Jensen K; Barzilay R Analyzing Learned Molecular Representations for Property Prediction. J. Chem. Inf. Model 2019, 59, 3370–3388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (34).Gilmer J; Schoenholz SS; Riley PF; Vinyals O; Dahl GE In Neural Message Passing for Quantum Chemistry. International Conference on Machine Learning, 2017; pp 1263–1272. [Google Scholar]
- (35).Swets JA Indices of discrimination or diagnostic accuracy. Psychol. Bull 1986, 99, 100–117. [PubMed] [Google Scholar]
- (36).Trott O; Olson AJ AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem 2009, 31, 455–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.