key: cord-0951008-9qtghr9o authors: Faquetti, Maria L.; Grisoni, Francesca; Schneider, Petra; Schneider, Gisbert; Burden, Andrea M. title: Identification of novel off targets of baricitinib and tofacitinib by machine learning with a focus on thrombosis and viral infection date: 2022-05-12 journal: Sci Rep DOI: 10.1038/s41598-022-11879-1 sha: 1f8824d0d1b7e1421030ecb0355699f6768900d3 doc_id: 951008 cord_uid: 9qtghr9o As there are no clear on-target mechanisms that explain the increased risk for thrombosis and viral infection or reactivation associated with JAK inhibitors, the observed elevated risk may be a result of an off-target effect. Computational approaches combined with in vitro studies can be used to predict and validate the potential for an approved drug to interact with additional (often unwanted) targets and identify potential safety-related concerns. Potential off-targets of the JAK inhibitors baricitinib and tofacitinib were identified using two established machine learning approaches based on ligand similarity. The identified targets related to thrombosis or viral infection/reactivation were subsequently validated using in vitro assays. Inhibitory activity was identified for four drug-target pairs (PDE10A [baricitinib], TRPM6 [tofacitinib], PKN2 [baricitinib, tofacitinib]). Previously unknown off-target interactions of the two JAK inhibitors were identified. As the proposed pharmacological effects of these interactions include attenuation of pulmonary vascular remodeling, modulation of HCV response, and hypomagnesemia, the newly identified off-target interactions cannot explain an increased risk of thrombosis or viral infection/reactivation. While further evidence is required to explain both the elevated thrombosis and viral infection/reactivation risk, our results add to the evidence that these JAK inhibitors are promiscuous binders and highlight the potential for repurposing. www.nature.com/scientificreports/ of all biological targets of a drug before its entrance into the market is often unfeasible. In this context, machine learning can be used to predict the potential for an approved drug to interact with off-targets and identify potential safety-related concerns 16 . The identification of additional drug-target interactions using chemo-centric and machine learning approaches and experimental confirmation may help to determine mechanisms of adverse drug events 17, 18 . For example, previously unknown drug-target interactions for the approved compound Celecoxib were identified using a ligand-based method, which is based on the principle that structural similarity reflects functional similarity, supporting the biological plausibility of reported cardiovascular adverse drug events 19, 20 . Moreover, off-target profiling is frequently used to identify candidate drugs for repurposing. For example, computational studies using machine learning identified baricitinib as a promising JAK inhibitor for repurposing in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 or COVID-19) 21, 22 . Baricitinib was considered a potential candidate for repurposing in COVID-19 based on the high affinity for AP-2 associated protein kinase 1 (AAK1) (EC number 2.7.11.1), which is critical in regulating viral endocytosis, and its inhibition may reduce the ability of the virus to infect lung cells 23 . Improving our understanding of the target space of JAK inhibitors drugs is essential in order to explain the mechanisms of unexpected side-effects associated with these drugs and to identify opportunities for repurposing. Although several binding screens of tofacitinib and baricitinib have been published, they are mostly limited to a few protein families, such as protein kinases and lipid kinases [24] [25] [26] . Thus, the community would benefit from a more extensive characterization of the target profile of tofacitinib and baricitinib. In light of the currently unexplained thrombotic and viral infection risk and the previously observed off-target binding potential of baricitinib, here we investigate if the thrombosis and viral infection/reactivation risk may be a result of an off-target effect. We, therefore, aimed to leverage well-established machine learning methods to identify off-target drug-protein interactions for baricitinib and tofacitinib and validate such predictions in vitro assays. Previously unknown off-targets of baricitinib and tofacitinib were predicted and confirmed drug-target interactions suggest an attenuation of pulmonary vascular remodeling, modulation of Hepatitis C (HCV) viral response, and hypomagnesemia. Nevertheless, the identified off-target interactions could not explain the elevated thrombosis or viral infections/reactivation risk. These results suggest both JAK inhibitors as potential candidates for drug repurposing. Off-target profiling of baricitinib and tofacitinib by machine learning revealed additional drug-target interactions. Macromolecular targets of baricitinib and tofacitinib were predicted using two previously published machine learning approaches: Target Inference Generator (TIGER) 20 and SOM-based Prediction of Drug Equivalence Relationships (SPiDER) 27 . Both approaches follow the chemical similarity principle, in which molecules sharing similar structures are likely to have similar bioactivity 28 . SPiDER uses a neural network (Self-organizing map [SOM]), and drug-target relationships are inferred based on descriptor similarity of a query compound to reference ligands without directly considering the target similarity. The method uses topological and physicochemical information of molecules to suggest a functional similarity between compounds. TIGER extends SPiDER using a more extensive set of targets, as well as a different prediction algorithm and scoring function. Both approaches have been extensively applied to de novo designed compounds, natural products with biological activity, and approved drugs [29] [30] [31] . Overall, 40 potential targets for baricitinib and 58 for tofacitinib (SPiDER [p < 0.05] and/or TIGER [score > 1]) were predicted by TIGER and SPiDER. The list of predicted targets accompanied by the score for TIGER, and p-values for SPiDER, are shown in Supplementary Tables S1 and S2, respectively. The cutoff values were chosen based on recent prospective studies where the target prediction tools led to bioactivity confirmation in vitro 20, 32, 33 . The resulting predictions reflect both known and unknown potential drug-target interactions. From all predicted targets, nine targets for baricitinib and eight for tofacitinib were identified as being relevant for thrombosis and viral infection/reactivation (Table 1) . www.nature.com/scientificreports/ In vitro characterization confirmed previously unknown baricitinib and tofacitinib drug-target interactions. Of the 98 predicted, a total of 11 drug-target interactions were experimentally validated using biochemical or cell-based assays, based on the availability of fee-based in vitro testing services (Table 2) . Among the predicted targets, two members of the Transient Receptor Potential superfamily of calcium channels were suggested, namely short transient receptor potential channels 6 (TRPC6) and 3 (TRPC3). Commercial assays were unavailable for TRPC6 or TRPC3, and therefore, these targets could not be tested. Instead, transient receptor potential cation channel subfamily M member 6 (TRPM6) (EC number 2.7.11.1) was employed for the respective binding assays. Additionally, while serine/threonine-protein kinase N2 (PKN2) (EC number 2.7.11.13) was Table 1 . Suggested targets with impact on thrombosis and viral infection per JAK inhibitor drug and target prediction approach. Commercial assays were unavailable for TRPC6 or TRPC3, and therefore, these targets could not be validated. Instead, transient receptor potential cation channel subfamily M member 6 (TRPM6) was employed for the respective binding assays. a PKN2 was included in the list of targets tested for baricitinib, which allowed us to make a direct comparison between tofacitinib and baricitinib inhibitory activity on this target. b Human herpesvirus 1 (strain SC16). www.nature.com/scientificreports/ among the predicted targets for tofacitinib, but not for baricitinib, PKN2-baricitinib binding affinity was previously determined in baricitinib (apparent dissociation constant [K d app ] = 269 nM and IC 50 = 284 nM) 25 . Thus, PKN2 was included in the list of targets tested for baricitinib, allowing a direct comparison between tofacitinib and baricitinib inhibitory activity on this target in the same experimental conditions. From the 11 drug-target interactions tested, five showed an experimental readout greater than 25% drugtarget interaction at 30 μM and were selected for further in vitro characterization ( Table 2 ). Four out of five drugtarget interactions were confirmed by further in vitro evaluation, with IC 50 and K i or K d values in the nanomolar range (baricitinib and tofacitinib on PKN2) and in the micromolar range (baricitinib on Phosphodiesterase 10A2 (PDE10A2) EC number 3.1.4.17; tofacitinib on TRPM6). The investigated drugs were considered as active if the determined IC 50 was lower than 30 μM. The raw in vitro data for drug-binding activity using biochemical assays is available in the Supplementary Table S3 . Dose-response curves for targets showing activity are available in the Supplementary Figs. S1-S4. Computational ligand docking predicted potential modes of baricitinib and tofacitinib-target interaction. Computational ligand docking (Fig. 2 ) predicted potential modes of interaction (i.e., threedimensional orientations of the drug molecule and a target) for baricitinib and tofacitinib in the binding pocket of the identified macromolecular targets (PKN2 [PDB-ID: 4CRS 34 ]; PDE10A [PDB-ID: 5C28 35 ]) using docking algorithms, and it provided the respective score for each orientation predicted. TRPM6 was not considered due to the unavailability of an experimentally determined structure. Molecular docking of baricitinib to PKN2, shown in Fig. 2a , suggests an interaction between the nitrile nitrogen on the drug structure and Mg 2+ . Similar to the crystallized ligand, the two residues Phee668 and Gly666, are hydrogen donors and interact with one oxygen from the sulfonyl group. Additionally, two arene-H interactions between pyrrole on the tofacitinib structure and the hydrogen on the amino groups of PKN2 residue Gly666 were suggested in Fig. 2b . A third arene-H interaction between pyrimidine on the tofacitinib molecule and the www.nature.com/scientificreports/ hydrogen on the amino groups of Lys686 was indicated. Like the crystallized ligand, an interaction with Phe668 is predicted. Additionally, interactions between Mg 2+ and two nitrogen atoms are suggested. In Fig. 2c , the Gln716 is making a hydrogen bond, donating a bond to the core pyrimidine nitrogen. Another interaction is suggested between the pyrimidine core of baricitinib and the protein-a π-stacking interaction with the key residue Phe719. Some structural equivalence between baricitinib and the co-crystallized ligand is observed, such as the pyrimidine core in the earliest and the aromatic ring in the latest. The interaction with Gln716 and Phe716 are key for recognition of PDE10A inhibitors by the enzyme 36 . Both baricitinib and tofacitinib were confirmed as "promiscuous drugs" since they bind to proteins from families other than the primary therapeutic target 20 . Thus, both drugs may be potential candidates for adverse drug effects and further repurposing. The confirmed drug-target interactions suggest an attenuation of pulmonary vascular remodeling (inhibition of PDE10A), modulation of Hepatitis C (HCV) viral response (inhibition of PKN2), and hypomagnesemia (inhibition of TRPM6). Therefore, we did not identify off-target interactions that could explain the elevated thrombosis or viral infections/reactivation risk observed in the clinical setting 5, 37, 38 . The thrombotic and cardiovascular risk associated with JAK inhibitors remains debated, which is largely due to a lack of a clear mechanism associated with the therapeutic target that could explain the increased risk. In our analysis, we aimed to investigate if there may be a plausible off-target interaction that could explain the observed effects. While the computational approaches identified several targets relevant for blood coagulation and platelet aggregation (e.g., Adenosine receptor A2A [AA2AR] and Arachidonate 15-lipoxygenase [15-ALOX] EC number 1.13.11.33), neither baricitinib nor tofacitinib was found to interact with those receptors in vitro, ruling them out as potential off-targets. Nonetheless, the drugs were shown to inhibit two targets related to thrombosis-PDE10A and TRPM6. PDE10A, which was recently validated as a novel target to treat pulmonary arterial hypertension (PAH) due to its central role in progressive pulmonary vascular remodeling 39, 40 , was identified as a target of baricitinib. The preliminary in vitro results of this study showed moderate inhibition of baricitinib for PDE10A2. Molecular docking in the active site of PDE10A (Fig. 2c) suggested a similar binding pose of baricitinib to the crystallized inhibitor (PDB ID: 5C28), with a predicted π-stacking interaction with the Phe719 residue, crucial for biological activity 36 . Additionally, among important regions for ligand binding is the occupation of a hydrophobic clamp formed by two phenylalanine residues, Phe719 and Phe686. The arene-H-type interaction between the pyrazole structure in baricitinib and Phe686 residue while occupying the hydrophobic clamp suggests that baricitinib has a similar binding mode to PQ-10, a papaverine analog having IC 50 equal to 6 μM. Clinically, PDE10A inhibition is expected to decrease the risk for thrombosis, particularly in patients with PAH. Thus, the expected positive clinical impact of PDE10A inhibition on the risk of thrombosis is not in line with a potential link to an elevated thrombosis risk. Rather, baricitinib might improve progressive pulmonary vascular remodeling. This study further identified previously unknown off-target interactions of tofacitinib with TRPM6, with moderate binding affinity. While our computational approach identified TRPC6 and TRPC3 as potential targets, we were unable to experimentally validate these targets due to a lack of commercially available in vitro assays. Thus, we can only speculate that the binding affinity observed with TRPM6 may translate to binding in TRPC6 and TRPC3. Additional experiments are needed to confirm if the C subfamily is also a potential target of tofacitinib. This aspect is important as TRPC6 is known to regulate human clot retraction, physiological hemostasis, and thrombus formation, and its inhibition is thought to have a positive effect on thrombotic outcomes 43 . Thus, further research is needed to confirm if tofacitinib binds to TRPC6. Cumulatively, the active targets in this study suggest that JAK inhibitors may have a beneficial effect on cardiovascular risk, and therefore do not support a hypothesis that the risk of thrombosis is related to an off-target drug effect (in the framework of the macromolecular targets investigated in this study). Nevertheless, we note that recent US-based cohort studies that have identified no difference in thrombosis risk between tofacitinib and TNF-inhibitors [44] [45] [46] , thereby suggesting that much of the observed risk seen in pharmacovigilance studies 6,47 may be due to underlying risk factors rather than a drug effect. For example, standardized incidence rates (IR) of venous thromboembolism or pulmonary embolism were comparable among patients with rheumatoid arthritis using tofacitinib (IR = 1.05 [0.78-1.39]) and bDMARDs (IR = 0.94 [0.85-1.03]) within MarketScan database cohorts 46 . Conversely, recent analysis using pharmacovigilance data of the US FDA Adverse Event Report System (FAERS) did not identify a signal of disproportionate reporting for venous thromboembolism and/or pulmonary embolism events with tofacitinib 46 . Therefore, an improved understanding of the underlying risk factors for thrombosis in patients with JAK inhibitors is urgently needed. The risk of thrombosis can be further increased in rheumatoid arthritis patients with high disease activity, cardiovascular risk factors (e.g., obesity), immobility, and hormonal replacement therapy 48, 49 . Patients using JAK inhibitors frequently have high disease activity with non-or inadequate response to csDMARDs and bDMARDs and multiple chronic conditions (e.g., cardiovascular disorders and depression), which can make the attribution of thrombotic events in patients treated with JAK inhibitors even more intricate. In addition to thrombosis, targets related to viral infection and viral reactivation were investigated. Therapies targeting the JAK family of enzymes may interfere with a normal antiviral response, including inhibition of IFN-γ activity, and may potentially increase the risk of infection/ reactivation of several viral infectious diseases, particularly HZ in rheumatoid arthritis patients 14, 38 . The computational approaches identified several targets expected to play a role in viral endocytosis and viral response, including epidermal growth factor receptor (EGFR) kinase (EC number 2.7.10.1) and PKN2. Although baricitinib and tofacitinib were not found to interact with EGFR in our experimental setting used, both showed PKN2 inhibitory activity. www.nature.com/scientificreports/ Clinically, PKN2 is of great importance as a target for antiviral therapy, particularly anti-HCV, as its suppression leads to viral replication blockage in humans. PKN2 inhibitors, in combination with other antiviral therapies, have demonstrated synergistic antiviral activity for chronic HCV treatment 50, 51 . While, to date, three studies have evaluated tofacitinib binding activity on PKN2, the results are contradictory 24, 25, 52 . The preliminary in vitro results of the current study suggest PKN2 inhibition with both baricitinib and tofacitinib, as the IC 50 and K i values are in the nanomolar range. Moreover, the molecular docking in the PKN2 crystalized structure suggested a similar binding mode, shape, and certain molecular features (i.e., pharmacophore) of baricitinib and tofacitinib (Fig. 1) as to the co-crystalized ligand at the protein binding site. The model indicates that the two drugs interact with the Mg 2+ similarly to the crystallized ligand-a PKN2 inhibitor-on the kinase functional pocket 53 . Similar to the crystallized ligand, each Phe668 and Gly666 as hydrogen donors and interact with one oxygen from the sulfonyl group in the baricitinib molecule, suggesting the role of this group for drug anchoring in the binding pocket. Additionally, the Phe668 residue backbone interacts with one nitrogen from the pyrimidine in tofacitinib, suggesting a similar binding mode to the crystallized ligand on the active pocket of PKN2. However, the impact of PKN2 inhibition is proposed to have a positive effect on viral suppression 51 , and therefore does not explain the elevated risk of HZ in rheumatoid arthritis patients. The exact mechanism of HZ viral reactivation remains unclear. Outside of its role in viral suppression, PKN2 may play an essential role in various cellular processes, such as cellular proliferation, migration, and signaling pathways [54] [55] [56] . Moreover, PKN2 is involved in autoinflammatory disorders 57 , heart failure 58 , and it is a target of interest in cancer 56, 59, 60 . As concerns regarding the risk of malignancy and major adverse cardiovascular events (MACE) in patients treated with tofacitinib have been raised by the European Medicines Agency, it is important to consider the potential role of PKN2 inhibition 61 , However, in mice models, PKN2 activation was the cause of cardiac dysfunctions 58 , and therefore, the clinical impact of PKN2 inhibition is contradictory to the risk of cancer and MACE in rheumatoid arthritis patients. Off-target profiling using computational approaches has been widely used to identify candidates for drug repurposing 62, 63 . Indeed, JAK inhibitors were recently established as potential candidate therapies for COVID-19 based on in silico methods [64] [65] [66] . Our computational methods identified 98 drug-target predictions, and the preliminary in vitro results found inhibitory activity on several proteins other than the primary therapeutic target, thereby confirming baricitinib and tofacitinib as promiscuous drugs and candidates for drug repurposing studies. For example, PDE10A inhibition has been primarily studied in psychiatric and neurological conditions, such as schizophrenia 67 and Huntington's disease 68 , and, to a lesser extent, in multiple peripheral pathological conditions 69,70 (e.g., osteogenic differentiation). Additionally, PDE10A inhibition by baricitinib is hypothesized to have a synergistic pharmacological effect in combination with other COVID-19 treatments (e.g., antiviral and corticosteroids drugs) due to the anti-fibrotic and anti-inflammatory effects of phosphodiesterase's inhibitors on the treatment of COVID-19 and its associated conditions (e.g., thrombosis, inflammation, and fibrosis) 71, 72 . Therefore, the confirmed PDE10A inhibition identified in this study supports the potential for baricitinib as a potential candidate outside of rheumatology. Moreover, while TRPM6 was not initially predicted, the moderate inhibitory activity is worth investigating. TRPM6 inhibition is not fully elucidated. However, it is mainly involved in magnesium homeostasis in the intestine and kidney 73, 74 , and it has been demonstrated to have expression levels modulated by hormones such as estrogen 75 and angiotensin II 76 , immunosuppressant 77 , and diuretics drugs 78 , and epidermal growth factor (EGF) 79 . Moreover, the decreased expression of TRPM6 in cancer patients treated with EGFR targeted therapies (e.g., cetuximab) seems to positively contribute to the oncologic efficacy of these therapies, as decreased magnesium availability inhibits cell proliferation and slows down tumor growth 79, 80 . Thus, we encourage further investigation into the clinical relevance of TRPM6 inhibition by tofacitinib in oncology. The results also highlight the complementarity of the two approaches TIGER and SPiDER. JAK3 (TIGER score = 6.9) and JAK1 (TIGER score = 1.9) ranked fourth and twenty-sixty, respectively, on the list of predicted target proteins for tofacitinib, while JAK3 (TIGER score = 8.4) ranked second for baricitinib using the TIGER approach. Tyrosine Kinase (EC number 2.7. Only a small fraction (~ 10%) of the 98 predicted off-targets were experimental tested in this study. However, as we did observe active binding on three distinct targets, this study suggests that there might be other interactions among the list of predicted targets. Thus, further testing might help to elucidate the molecular mechanisms of these JAK inhibitors and open the door for improved understanding of the safety concerns and repurposing in other conditions (e.g., in neurodegenerative diseases, diabetes, and viral infections). The use of computational and experimental approaches in this study allowed for the identification and characterization of previously unknown off-target interactions for the two JAK inhibitors (e.g., baricitinib-PDE10A and tofacitinib-TRPM6), which adds to the target space of tofacitinib and baricitinib. TIGER and SPiDER identified additional targets of tofacitinib and baricitinib that other approaches, such as the Similarity Ensemble Approach (SEA) 81 www.nature.com/scientificreports/ be performed with caution, as extensive experimental studies are essential for validating the hypotheses and demonstrating the potential impact of each approach. Moreover, TIGER and SPiDER use a large set of targets, encompassing a broad scope of protein families that allows identifying drug promiscuity. Additionally, the inclusion of multiple protein families helps to predict a broad off-target drug profile and point out potential targets for repurposing studies. This is particularly important for understudied druggable proteins and targets with no approved drugs. Ultimately, it increases knowledge on the potential drug effects of tofacitinib and baricitinib. Despite the encouraging results of our study, we are mindful of some limitations. As identified, we could not experimentally validate all predicted targets related to thrombosis (e.g., TRPC6) . As such, we cannot conclude if these targets may play an important role in thrombosis or viral infection/reactivation risk and are limited in the conclusions we can draw. Thus, we encourage researchers with access to the appropriate assays to validate these targets. Moreover, there might be additional targets of relevance that were not predicted by our computational tools. The provided docking poses constitute an additional support to the experimentally determined values and that shall not be considered as a binding hypothesis. Thus, future computational studies including X-ray crystallography analysis are needed, and it may provide insights on the binding mode of the JAK inhibitors on the new targets. We also acknowledge that the activity of small molecule drugs using in vitro assays does not always translate into activity in the cellular environment. Thus, the results should still be interpreted with caution and treated as preliminary evidence for the off-target binding of baricitinib and tofacitinib. In summary, previously unknown off-targets of baricitinib and tofacitinib were identified and characterized using a combination of machine learning and experimental methods. The confirmed target interactions suggest an attenuation of pulmonary vascular remodeling, modulation of HCV viral response, and hypomagnesemia. Thus, it does not endorse the hypothesis of elevated thrombosis or viral infections/reactivation risk explained by one (or more) drug-target interactions. Consequently, the current safety concerns may be due to underlying patient-specific factors (confounders) or to targets not detected by our computational pipeline. Additionally, as not all of the predicted targets were experimentally validated, further research is warranted. Finally, baricitinib and tofacitinib may be potential candidates for repurposing, as they were identified as drugs with promiscuous binding activity. Data preparation and molecular representation for target prediction. Baricitinib and tofacitinib were provided as Simplified Molecular Input Line Entry Specification (SMILES) and processed in KNIME v3.7.2 83 with the MOE v.2019.0102 84 "wash" function employing the following options: "disconnect salts", "remove lone pairs", "deprotonate strong acids", "remove minor component", "protonate strong bases, " and "add hydrogen". Chemically advanced template search version 2 (CATS2) 85 descriptors and two-dimensional MOE descriptors ('QSAR descriptors' node of KNIME; 'Forcefield' = MMFF94*) were calculated for all generated molecules and used as input for the target prediction tools; Macromolecular target prediction and selection. Target Inference Generator ([TIGER v. 19 .07], inSili.com. LLC, Zurich) 20 and Self-organizing map-based prediction of drug equivalence relationships (SPi-DER) 27 software's were used for target activity prediction. Targets with statistically meaningful predictions from SPiDER (p < 0.05) and/or TIGER (score > 1) were selected for in vitro characterization if they were considered to have a potential influence on thrombosis or viral infection/reactivation. In vitro characterization. Baricitinib (99.97% purity) and tofacitinib (99.96% purity) compounds were purchased from MedChemExpress LLC (New Jersey, www. medch emexp ress. com). In vitro characterization was performed on a fee-for-service basis at Eurofins (www. eurofi ns. com) if the assay was commercially available. For the biochemical assays, compound targets showing an experimental readout greater than 25% (inhibition or stimulation) at 30 µM were selected for follow-up, and dose-response curve characterization and determination IC 50 /EC 50 (two or three replicates, multiple concentrations, maximum 100 µM concentration). Additional details on the conduct of in vitro assays are included in Supplementary Information S1. www.nature.com/scientificreports/ ester in the binding site of PKN2 led to led to a RMSD value of 0.80 Å for 4CRS, while re-docking of 6-chloro-2-cyclopropyl-5-methylpyrimidin-4-amine in the binding site of PDE10A led to a value of RMSD = 0.23 Å for 5C28. JAK inhibitors were docked into the crystalized structure (baricitinib and tofacitinib using GoldScore scoring function on PKN2 [PDB ID: 4CRS], and baricitinib using PLP scoring function on PDE10A [PDB ID: 5C28]) and the minimum energy pose was chosen for the analysis. Statistical analysis. Transient receptor potential cation channel subfamily M member 6 (TRPM6). The binding constant (K d ) was calculated with a standard dose-response curve using the Hill Eq. (1): The curve was fitted using a non-linear least square fit with the Levenberg-Marquardt algorithm using RStudio Team (2020) All data generated or analyzed during this study are included in this published article (and its Supplementary Information files S1). Any additional materials will be provided upon request. Requests for the materials should be submitted to A.M. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological diseasemodifying antirheumatic drugs: 2019 update JAKS and STATS: Biological implications Targeting the JAK/STAT signaling pathway for breast cancer SARS-CoV-2 induced neurological manifestations entangles cytokine storm that implicates for therapeutic strategies Thromboembolism with Janus Kinase (JAK) inhibitors for rheumatoid arthritis: How real is the risk? Analysis of spontaneous postmarket case reports submitted to the FDA regarding thromboembolic adverse events and JAK inhibitors Real-world comparative risks of herpes virus infections in tofacitinib and biologic-treated patients with rheumatoid arthritis FDA approves Boxed Warning about increased risk of blood clots and death with higher dose of arthritis and ulcerative colitis medicine tofacitinib (Xeljanz, Xeljanz XR). US Food Drug Adm Increased risk of blood clots in lungs and death with higher dose Xeljanz (tofacitinib) for rheumatoid arthritis Olumiant (baricitinib) FDA Approval History European Medicines Agency JAK inhibition as a therapeutic strategy for immune and inflammatory diseases Herpes zoster and tofacitinib therapy in patients with rheumatoid arthritis Herpes Zoster and Tofacitinib: Clinical Outcomes and the Risk of Concomitant Therapy Polypharmacology in drug discovery Predicting new molecular targets for known drugs Application of machine learning techniques for drug discovery Building Machine-Learning Scoring Functions for Structure-Based Prediction of Intermolecular Binding Affinity Computational approaches in chemogenomics and chemical biology: Current and future impact on drug discovery A computational method for unveiling the target promiscuity of pharmacologically active compounds Baricitinib as potential treatment for 2019-nCoV acute respiratory disease Planning chemical syntheses with deep neural networks and symbolic AI Genomic characterisation and epidemiology of 2019 novel coronavirus: Implications for virus origins and receptor binding A quantitative analysis of kinase inhibitor selectivity The target landscape of clinical kinase drugs Chemical proteomics reveals target selectivity of clinical Jak inhibitors in human primary cells Identifying the macromolecular targets of de novo-designed chemical entities through self-organizing map consensus Molecular similarity in medicinal chemistry Spotting and designing promiscuous ligands for drug discovery Design of natural-product-inspired multitarget ligands by machine learning Polypharmacological drug−target inference for chemogenomics Synthetic activators of cell migration designed by constructive machine learning Computer-assisted discovery of retinoid x receptor modulating natural products and isofunctional mimetics Human protein kinase N2 (PKN2, PRKCL2) in complex with ATPgammaS. Worldwide Protein Data Bank Discovery and optimization of a series of pyrimidine-based phosphodiesterase 10A (PDE10A) inhibitors through fragment screening, structure-based design, and parallel synthesis Impact of Janus kinase inhibitors on risk of cardiovascular events in patients with rheumatoid arthritis: systematic review and meta-analysis of randomised controlled trials A systematic review and meta-analysis of infection risk with small molecule JAK inhibitors in rheumatoid arthritis Validation of phosphodiesterase-10 as a novel target for pulmonary arterial hypertension via highly selective and subnanomolar inhibitors Phosphodiesterase 10A upregulation contributes to pulmonary vascular remodeling Discovery of a series of 6,7-dimethoxy-4-pyrrolidylquinazoline PDE10A inhibitors Current Landscape of Phosphodiesterase 10A (PDE10A) Inhibition A critical role for the transient receptor potential channel type 6 in human platelet activation Comparative risk of venous thromboembolism in rheumatoid arthritis patients receiving tofacitinib versus those receiving tumor necrosis factor inhibitors: An observational cohort study Risk of venous thromboembolism associated with tofacitinib in patients with rheumatoid arthritis: a population-based cohort study. Rheumatology (Oxford). keab29 Incidence of venous and arterial thromboembolic events reported in the tofacitinib rheumatoid arthritis, psoriasis and psoriatic arthritis development programmes and from real-world data Thromboembolic safety reporting of tofacitinib and baricitinib: An analysis of the WHO VigiBase Risk of venous thromboembolism in rheumatoid arthritis, and its association with disease activity: a nationwide cohort study from Sweden Epidemiology and risk factors for venous thrombosis HA1077 displays synergistic activity with daclatasvir against hepatitis C virus and suppresses the emergence of NS5A resistance-associated substitutions in mice Suppression of hepatitis C virus replication by protein kinase C-related kinase 2 inhibitors that block phosphorylation of viral RNA polymerase Comprehensive analysis of kinase inhibitor selectivity Purification and kinase assay of PKN Rho GTPases regulate PRK2/PKN2 to control entry into mitosis and exit from cytokinesis Regulatory domain selectivity in the cell-type specific PKN-dependence of cell migration Protein kinase C inhibitor chelerythrine selectively inhibits proliferation of triple-negative breast cancer cells Pyrin inflammasome activation and RhoA signaling in the autoinflammatory diseases FMF and HIDS Protein kinase N promotes stress-induced cardiac dysfunction through phosphorylation of myocardin-related transcription factor A and disruption of its interaction with actin Protein kinase C-related kinase 1 and 2 play an essential role in thromboxane-mediated neoplastic responses in prostate cancer Role of protein kinase N2 (PKN2) in cigarette smoke-mediated oncogenic transformation of oral cells Pfizer shares co-primary endpoint results from post-marketing required safety study of XELJANZ® (tofacitinib) in subjects with rheumatoid arthritis (RA) New uses for old drugs Associating drugs, targets and clinical outcomes into an integrated network affords a new platform for computeraided drug repurposing Potential role of Janus kinase inhibitors in COVID-19 HiJAKing SARS-CoV-2? The potential role of JAK inhibitors in the management of COVID-19 Mechanism of baricitinib supports artificial intelligence-predicted testing in COVID-19 patients Use of structure-based design to discover a potent, selective, in vivo active phosphodiesterase 10A inhibitor lead series for the treatment of schizophrenia Chronic suppression of phosphodiesterase 10A alters striatal expression of genes responsible for neurotransmitter synthesis, neurotransmission, and signaling pathways implicated in Huntington's disease A novel role of cyclic nucleotide phosphodiesterase 10A in pathological cardiac remodeling and dysfunction Phosphodiesterase 10A is a mediator of osteogenic differentiation and mechanotransduction in bone marrow-derived mesenchymal stromal cells Phosphodiesterase inhibitors: could they be beneficial for the treatment of COVID-19? Commentary: Phosphodiesterase 4 inhibitors as potential adjunct treatment targeting the cytokine storm in COVID-19 Role of kinase-coupled TRP channels in mineral homeostasis TRPM6 and TRPM7-Gatekeepers of human magnesium metabolism Differential regulation of transient receptor potential melastatin 6 and 7 cation channels by ANG II in vascular smooth muscle cells from spontaneously hypertensive rats Downregulation of Ca2+ and Mg2+ transport proteins in the kidney explains tacrolimus (FK506)-induced hypercalciuria and hypomagnesemia Enhanced passive Ca2+ reabsorption and reduced Mg2+ channel abundance explains thiazide-induced hypocalciuria and hypomagnesemia Impaired basolateral sorting of pro-EGF causes isolated recessive renal hypomagnesemia Early magnesium reduction in advanced colorectal cancer patients treated with cetuximab plus irinotecan as predictive factor of efficacy and outcome Magnesium deficiency inhibits primary tumor growth but favors metastasis in mice Relating protein pharmacology by ligand chemistry SwissTargetPrediction: updated data and new features for efficient prediction of protein targets of small molecules KNIME: The Konstanz Information Miner Molecular Operating Environment (MOE). (Chemical Computing Group ULC Chemically Advanced Template Search (CATS) for Scaffold-Hopping and Prospective Target Prediction for 'Orphan' Development and validation of a genetic algorithm for flexible docking Relationship between the inhibition constant (KI) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction We thank Dr. Jan A. Hiss, Sarah Haller, and Dr. Stefanie-Dorothea Krämer for technical support. This article was funded by the Eidgenössische Technische Hochschule (ETH) Zürich Foundation (COVID-19 research grant), and the salary of M.L.F. was supported by an ETH Grant (ETH-32 18-2). PS and GS declare a potential financial conflict of interest as founders of inSili.com GmbH, Zurich, and in their role as consultants to the pharmaceutical industry. The professorship of AMB was partially endowed by the ETH Foundation and PharmaSuisse. MLF and FG declare that they have no competing interests. The online version contains supplementary material available at https:// doi. org/ 10. 1038/ s41598-022-11879-1.Correspondence and requests for materials should be addressed to A.M.B.Reprints and permissions information is available at www.nature.com/reprints.Publisher's note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.