Skip to main content
Neurotherapeutics logoLink to Neurotherapeutics
. 2025 Jun 17;22(4):e00624. doi: 10.1016/j.neurot.2025.e00624

Applications of artificial intelligence in drug discovery for neurological diseases

Sean Ekins 1,, Thomas R Lane 1
PMCID: PMC12418477  PMID: 40533306

Abstract

Neurological disease encompasses over 1000 disorders, exacts a massive human health and financial toll as well as being a story of extremes. At one end are diseases that are complex and heterogeneous affecting millions, while at the other there are monogenic and rare diseases, with a handful of individuals. What are absent are drugs that can treat or cure the disease. Discovering these is challenging, held back by extreme costs to develop them or in some cases by the limited understanding of the diseases. After decades of drug discovery research there is now considerable data available which can be used to help develop novel compounds more strategically. This includes high throughput screening data with targets, crystal structures of proteins implicated in neurological diseases and adjacent data such as properties of molecules like blood brain barrier permeability as well as an array of in vitro and in vivo toxicity endpoints valuable for any drug targeting the central nervous system. While computational tools have been developing and applied to neurological diseases for decades, we are now in the age of machine learning and artificial intelligence (AI). This promises the potential to expedite the identification and discovery of new molecules. Whether by using individual computational techniques or complex end-to-end approaches, scientists can narrow the molecules they make and test as well as study more targets or diseases which might have been out of reach previously. This review highlights the many different applications of AI potentially enabling new discoveries and treatments for neurological diseases.

Keywords: Artificial intelligence, Machine learning, Neurological diseases

Graphical abstract

Image 1

Introduction

Over the last 30 years, we have seen big pharmaceutical companies go through cycles in which they were productive in discovering new classes of drugs targeting the brain (1990s), and then not long after a period in which productivity declined (2000s) and many exited neurological disease research. Presently we are seeing a renewed interest. Why is this? Have there been any new breakthroughs in the science, are they chasing new financial opportunities or is it a symptom of “what is old is new again”? With each passing decade developing new pharmaceuticals has become increasingly expensive with staggering numbers in recent years (from $1bn [[1], [2], [3]] to ​> ​$4bn [4]). This cost can only be partially attributed to the high attrition rate with only a 14 ​% success rate of moving compounds from the clinic to the market between 2006 and 2022 [5]. Of particular note central nervous system (CNS) drugs suffer from the highest attrition rate, with only ∼7 ​% ultimately reaching the marketplace after an average of 12.6 years in development [6]. Developing drugs that target the brain for neurological indications is therefore certainly a difficult challenge and not one for the faint of heart. One example of a complex neurodegenerative disease is Alzheimer's disease (AD), which affects 10's of millions of individuals, with the underlying mechanisms of its pathophysiology still unclear. There are also rare diseases that affect a handful of patients caused by a mutation in a single (likely essential) gene which may have a devastating effect on development and shorten the life of the patient (e.g. Batten disease CLN1 as an example [7]). In both examples, to get to a reliable treatment, one must get the drug into the brain, then deliver the small molecule or biologic to the desired target receptor or organelle in the right region/s. This is akin to addressing a letter with a full address and zip code to ensure the exact delivery to the desired location. Optimal drug discovery for both common and rare neurological diseases would involve the ability to predict key factors across the development pipeline, from designing molecules with optimal target engagement and physicochemical properties for oral bioavailability and blood–brain barrier (BBB) penetration, to anticipating clinical performance in humans [8]. This serves as a prelude to a review of artificial intelligence (AI) applications in neurological drug discovery, drawing on both our work and that of others. Firstly, we present a brief rationale for the need of such approaches.

The need for treatments, the cost of neurological diseases and the opportunity

There is no doubt that neurological diseases inflict an incredible cost on 100's millions of patients in society. Over the years we have seen some quite staggering statistics and numbers which were likely already outdated by the time they were published. For example, AD is an irreversible neurodegenerative disease widely known as the most common cause of dementia, affects over 5.1 million adults over age 65, costs over $150 billion per year [9] and is the 6th leading cause of death in USA. AD is a typical age-dependent neurodegenerative disease that affects 5 ​% of individuals >65 years, 20 ​% of those >85 years, and more than one-third of those >90 years [10]. However, approximately 5 ​% of the 6.5 million people in the United States have younger-onset AD diagnosed under age 65. The mechanisms underlying AD pathophysiology are still unclear. Aggregation of tau and amyloid beta (Aβ) proteins as well as decreased acetylcholine (ACh) are the focus of many studies [11]. To date several in vivo models of AD have shown some improvement in cognition as well as tau and Aβ pathology [11]. While there are recently approved biologics (e.g. aducanumab and lecanemab-irmb), the only small molecule therapies that are currently on the market for treatment of AD are four acetylcholinesterase (AChE) inhibitors (tacrine, rivastigmine, donepezil and galantamine) and one N-methyl-d-aspartate (NMDA) receptor inhibitor (memantine) which are only symptomatic treatments and do not affect the underlying disease mechanisms or alter the disease course [9]. So, there is a huge opportunity to address this unmet need for AD.

Another important neurological disease is epilepsy, which affects around 50 million people worldwide, accounting for one third of the world's neurological disease burden [12,13]. The World Health Organization (WHO) estimated that 5 million people are diagnosed with epilepsy each year globally. The cause of epilepsy is still unknown in about 50 ​% of cases, although several disease mechanisms may lead to epilepsy. Critically, toxicity is one of the most important reasons for the failure of most clinical studies, with CNS toxicity linked to a 30 ​% clinical trial failure rate [14]. One of the more common drug-induced CNS issues is seizure liability, which leads to many challenges including reduced competitiveness, project delays, and increased costs of developing these drugs [15,16].

A major focus of many CNS targeting drugs is treating acute and or chronic pain. A study in 2011 by the Institute of Medicine found that chronic pain affects at least 100 million adults and costs society > $560 billion annually in the USA alone. Decades of overreliance on opioids such as fentanyl for treating chronic pain (despite their poor ability to improve function) has contributed to an epidemic of opioid overdose deaths and addictions in the US. The total number of synthetic opioid-related deaths in recent years has hovered between ∼74,000–76,000 according to the National Center for Health Statistics at the CDC [17]. One study recently estimated the annual financial cost to be around $504 billion [18]. Despite the enormous cost of opioid use disorder (OUD) in the United States and world-wide, there has been limited success in the creation of treatments. These examples may just be the tip of the iceberg in terms of a small selection of neurological diseases, but cumulatively the numbers of patients and costs to society of are collectively immense. If we remember that there are over 1000 neurological disorders (common and rare) [19], hence finding ways to develop treatments for all of these is a herculean challenge for any technology, but in recent years machine learning and AI approaches have appeared from disparate areas that may provide some hope or at least a vision of what may be possible in future.

Introduction to machine learning and AI

One way to differentiate machine learning from artificial intelligence (Fig. 1) is to remember the former learns from data and makes decisions on that within a defined set of rules and does not create anything new. AI however can learn from the data and make decisions based on it that to go beyond the training data so that it can create something new or optimize an outcome. When we build a machine learning model, we are generally using one of many potential algorithms to pick up the pattern in the properties in the dataset that relates to a measured output of interest. For example, with a set of small molecules we can generate potentially thousands of different types of molecule descriptors (features that describe the 1D, 2D, 3D properties or features of molecules) which we can then use with an algorithm (e.g. support vector machine, random forest, deep learning or another) to either classify the measured activity related to the data or to generate a regression model (to predict a continuous value). There are many examples in the following sections on different types of data that are modeled relating to neurological diseases, although this is dominated by in vitro data due to the increasing quantity available. There are far fewer examples of true AI applications were something new is created like a molecule for a specific protein involved in a neurological disease, likely because these techniques are at an earlier stage of technological development and validation.

Fig. 1.

Fig. 1

Differentiating traditional AI (machine learning) from Generative AI.

Finding molecules for neurological diseases using AI

An often-used analogy in drug discovery, is that it is like finding a needle in a haystack. If you are looking for a small molecule there are billions of possibilities [20], so narrowing it down to a manageable number to make and test would be a viable starting point that would be financially feasible without breaking the bank. There are several ways to accelerate this search that are commonly used. One would be to take a target implicated in a neurological disease and dock millions of molecules into the binding site to filter them down to a set to ultimately test in vitro before in turn selecting the best to test in vivo. Of course, until recently if there was no crystal structure of a target you would need to generate a homology model. AlphaFold [21] has changed this using AI technology literally overnight, and this has greatly improved the numbers and quality of such models of human proteins available (in most cases). As an example, a recent study docked more than 16 million compounds in models derived from AlphaFold and other homology models for the trace amine-associated receptor 1 (TAAR1) [22]. From 62 molecules that were purchased and tested, 25 were agonists in in vitro [22]. In the absence of the target structure, one could also generate a machine learning model that learns from experimental data (such as Ki, IC50, EC50). In ChEMBL, there is data for over 600 agonists which after curation and cleaning can be used to generate a machine learning models for TAAR1 as just one example (Fig. 2). While a comparison of docking to ligand-based machine learning and in turn HTS approaches was not undertaken, this kind of prospective approach needs to be performed to provide some confidence in what these technologies can do in isolation or when combined. What follows are several examples of using ligand-based machine learning models to find new molecules for various targets (Table 1). It should however be noted that as with other areas there is likely substantial publication bias of positive examples and demonstrations of ‘failures’ with these methods are rarely if ever published.

Fig. 2.

Fig. 2

5-fold cross validation metrics for classification (A) and regression (B) models for human TAAR1 agonism using the support vector machine classification (svc) and regression (svr) algorithms. Training data was obtained from ChEMBL and filtered to only include those with endpoints associated with agonism. (A) Classification models include a histogram of the probability-like score distributions (x-axis) with red and blue colors representing the active and inactive ground truth, respectively. (B) Regression model cross validation shows the actual (x-axis) versus the predicted (y-axis) activity (-logM).

Table 1.

Machine learning model statistics for neurological targets. B, Bayesian; DL, deep learning; KNN, K-nearest neighbors; LREG, logistic regression; RF, random forest; SVC, support vector classification, SVR, support vector regression; XGB, XGBoost.

Target N 5- fold AUC Model type External validation Reference
ACHE Eel 4545
Human 4601
Eel 5459
Human 4075
Rat 1406
Mouse 368
Cow 457
Ray 307
Mosquito 72
0.92
0.91
0.94
0.94
0.95
0.95
0.94
0.95
1
B
B
RF
RF, SVC
DL
DL, RF
DL, LREG, RF
DL, SVC
DL
66 molecules tested
N ​= ​203 - AUC 0.90
N ​= ​208 - AUC 0.82
[46]
[46]
[49]
[49]
[49]
[49]
[49]
[49]
[49]
BCHE 2496 0.94 Bayesian [46]
CCR3 699 0.94 RF, SVC, XGB 17 molecules tested [82]
CCR4 1273 0.98 KNN, LREG, DL, RF, SVC, XGB 17 molecules tested [82]
CCR5 1948 0.94 SVC [82]
P2Y6R 244 0.79 RF, XGB 19 compounds tested [104]
A1AR 132 0.87 B 29 compounds tested [112]
GSK-3β 2368
2618
0.90
0.85
B
SVR
5 compounds tested
Tested with an external dataset
[147]
[148]
Nav 1.8 1276 0.84 B [164]
Kv7.1 1276 0.77 B [164]

Enzyme machine learning models

AChE has long been considered a key therapeutic target in the symptomatic treatment of AD to treat severe cognitive deficiency [23]. AChE inhibitors have also been implicated in the treatment of many other neurological diseases including Parkinson's disease, other form of dementias and autoimmune disorders like Myasthenia Gravis [24,25]. Inhibitors of this enzyme work by reversibly blocking binding of the substrate to AChE or by hydrolytic inactivation of AChE, resulting in an increasing concentration of the neurotransmitter acetylcholine (ACh) at the synapse. Traditional synthetic chemistry efforts employing medicinal chemistry have been a fertile source of multiple classes of AChE inhibitors over the years [26], which in turn have been employed to build a wide array of quantitative structure activity relationship (QSAR) models, machine learning models [[27], [28], [29], [30], [31], [32], [33], [34]] or protein based-docking models [35]. Many computational structure-based pharmacophore modeling [36], machine learning [37], molecular docking [[38], [39], [40]], 2D and 3D similarity searches [41], MIA-QSAR modeling [42], or combinations of these different strategies [[43], [44], [45]] have also been used. This also highlights that there is considerable in vitro data to model for generally narrow chemical series, but cumulatively there is also sufficient data for machine learning.

We have demonstrated this by developing Bayesian and other machine learning models for eel and human AChE using data from ChEMBL for over 4000 molecules [46]. We have observed very similar 5-fold cross validation model statistics across machine learning models for both species. These models were also used to search the MicroSource Spectrum collection (2431 compounds) to identify novel AChE inhibitors. 66 compounds were predicted as “active” against eel AChE (inhibiting enzyme activity ≥50 ​% at 20 ​μM). Seven of these compounds were identified as novel eel AChE inhibitors including tilorone (IC50 of 14.4 ​nM) for the eel acetylcholinesterase. Tilorone also showed an IC50 of 73.3 ​nM against the human AChE and was verified by a secondary assay (IC50 of 56 ​nM) [46].

Following this study we further curated AChE inhibition data from public sources for many other species using data from ChEMBL [47] and BindingDB [48]. For each species we built 8 machine learning models using our Assay Central software and validated them by identifying published molecules that inhibit human AChE [49]. As there are many other different machine learning approaches available, we also performed a comparison of our descriptor-based models against a more recent machine learning algorithm called AttentiveFP [50] using the larger eel and human datasets. The internal cross-validation statistics of these models was performed using nested, five-fold cross validation (apart from DL, which used a 20 ​% leave out set), and for AttentiveFP which used five-fold cross validation. These 5-fold cross validation statistics for the different AChE models had AUC and F1 scores >0.9. To further test the utility of the models developed we used external validation for the human and eel models performed using test sets that had been manually curated from the literature for additional inhibitors of human and eel AChE. As we had generated many different types of models we predicted these test sets against consensus models for each species and ultimately demonstrated that the human and eel consensus models could predict activity in the test sets with 81 ​% and 82 ​% accuracy, respectively. We also illustrated that although the models had good predictions for general AChE inhibition, there was no species specificity. Besides classification models, regression models were also developed for the human and eel datasets which were of a smaller size. We found that the SVR algorithm outperformed all the other algorithms for both human and eel with R2 values of 0.81 and 0.75, respectively. These regression models were subsequently used to predict the potency of the compounds in the human and eel external test sets and the human test set predicted with the human regression SVR model performed the best (Pearson's r ​= ​0.76, mean absolute percentage error ​= ​9.73 ​%). In order to perform prospective prediction these models were used to score >195,000 compounds using a larger human AChE model machine learning model consisting of over 10,000 molecules. This resulted in 111 compounds predicted to be active with an applicability domain score ≥0.8. From these we tested ten compounds for potential AChE inhibitory activity and eight of the ten selected compounds displayed modest inhibition of AChE at 100 ​μM, (with six of the ten displaying >10 ​% activity at this concentration and 2 of the that IC50's of between 7 and 14 ​μM). This demonstrated that the machine learning model virtual screening could suggest new human AChE inhibitors [49]. In contrast to the dogma around quality of literature data derived from different laboratories, we have demonstrated how careful curation of such data can be used to develop machine learning models with value in finding new molecules with biological activity.

A closely related enzyme is butyrylcholinesterase (BChE) which is a hydrolase responsible for hydrolyzing esters of choline including ACh [51,52], but with less efficiency than AChE [53]. Accumulating evidence indicates that BChE may have neuronal and non-neuronal roles in the CNS due to the wide distribution of neuronal BChE in certain brain regions [54,55]. It is widely known that degeneration of the basal forebrain cholinergic system is an indication of AD [56]. Studies have found that BChE biochemical properties were changed in neurodegenerative diseases like in AD. For example, in the cortical region the BChE level is increased during AD which is responsible for plaques and neurofibrillary tangle formation [56]. Additionally the inhibition of BChE results in an increase in the ACh levels in the brain, which further suggests a regulatory function in the hydrolysis of ACh [57]. BChE is also involved in the regulation and development of the nervous system in several species [[58], [59], [60], [61]]. BChE inhibitors have therefore been suggested as a treatment for AD in which the AChE-to-BChE ratio changes [62,63] and would additionally counter the side effects of AChE inhibitors such as their gastointestinal and hepatotoxic effects [[64], [65], [66]] 18. The identification of selective BChE inhibitors has lagged behind AChE research but has become attractive, and remains challenging due to the structural similarities between AChE and BChE, (65 ​% amino acid sequence homology [53,54,[67], [68], [69]] and similar catalytic active site, gorge structure and peripheral anionic site [67,69,70]). We have therefore investigated the use of several machine learning algorithms and different model training procedures in an effort to identify selective BChE inhibitors in a large library of compounds. As described previously human AChE and additionally BChE inhibition data (IC50) were acquired from CHEMBL [49] with only compounds with inhibition data for both targets included in the training dataset [71]. Several machine learning algorithms, including the state-of-the-art contrastive learning (CL) were used to identify selective BChE inhibitors. CL is a machine learning algorithm that was originally developed to label data without supervision [72,73], followed by a supervised version [74] of the same learning schema to generate a more accurate representation of data points and hence provide improved classification accuracy [71]. In addition to the supervised CL, we tested a deep learning and random forest model for comparison. The 3 algorithms were used to generate many models and predict a vendor library consisting of ∼5 million compounds. After filtering with absorption, distribution, metabolism, excretion and toxicity (ADME/Tox) models we shortlisted 30 compounds in total (12 from the DL model, 11 from the CL model and 7 from the RF model). 20 compounds were purchased and tested in vitro using both human BChE and AChE assays. Ultimately we identified 7 promising selective BChE inhibitors which did not inhibit AChE appreciably [71].

GPCR machine learning models

G protein-coupled receptors (GPCRs) are essentially sensors that enable the nervous system to respond to external stimuli [75]. They have very similar 7 transmembrane alpha helices and can engage different types of molecules of varying sizes. 35 ​% of FDA approved drugs act via these GPCR proteins and are linked with many major neurodegenerative diseases (such as AD, Parkinson's disease (PD), Huntington's (HD), depression, attention deficit hyperactivity disorder (ADHD), schizophrenia etc.). An excellent recent 2023 review by Wong et al. [75] describes the extensive role of GPCRs in these neurodegenerative diseases, although it did not delve into how computational, or AI approaches could be used to identify ligands for these receptors for neurodegenerative diseases. An earlier review on computational approaches in GPCR ligand discovery had highlighted structure-based design and ligand-based approaches and workflows with some examples related to CNS targets such as the dopamine D2 receptor, D4 receptor, mu-opioid receptor 5-HT1A, 5-HT1A 5-HT2A, 5-HT2B, and adenosine A1A, A2A, and purinergic P2Y receptors [76] with only limited examples of molecules that were described as “in clinical trials”. What follows in this section are several examples of applications of machine learning and AI to GPCRs (Table 1).

Chemokine receptors are G protein-coupled receptors implicated in numerous diseases [[77], [78], [79]]. Recently, CCR3 and CCR4 have emerged as potential stroke targets as these genes are predictors of both infarct volume and edema in stroke patients undergoing thrombectomy [80]. In addition, blocking multiple chemokine receptors was suggested as a potential treatment for neuropathic pain alongside opioids [81]. We have curated data for CCR3, CCR4, and CCR5 antagonists from the ChEMBL database and used various machine learning models for virtual screening, followed by testing 17 compounds in vitro [82]. In the process we demonstrated the repurposing of the FDA approved dual tyrosine kinase inhibitor lapatinib which had in vitro activity against all three CCRs [82]. CCR5 has also recently been implicated as a target in stroke and traumatic brain injury by others [83].

Modulators of purinergic signaling including 12 GPCRs (8 P2Y and 4 adenosine receptors) are relevant to many pathological conditions and therapeutic development [84], with the Gq protein-coupled P2Y6 receptor (P2Y6R) being one of the most important targets involved in cell proliferation, inflammation, cerebroprotection and immune responses [85,86]. The activation of the P2Y6R has been linked to AD, PD, epilepsy, chronic neuropathic pain and other diseases [85,[87], [88], [89], [90], [91], [92], [93], [94]] yet there are few selective P2Y6R antagonists although there has been considerable research [[95], [96], [97], [98]]. This research has focused on different classes of compounds such as diisothiocyanate derivatives [99,100], chromene derivatives [101,102] and 2-(1-(tert-butyl)-5-(furan-2-yl)-1H-pyrazol-3-yl)-1H-benzo [d]imidazole derivatives [103]. We have used public data for P2Y6R agonists to develop machine learning models and validated these classification models through in vitro testing (human P2Y6R and hP2Y14R) [104]. Compounds identified included ABBV-744, an experimental anticancer drug which inhibited hP2Y6R and showed hP2Y1R enhancement. Other compounds identified included the experimental anti-asthmatic drug AZD5423, as well as TAK-593 and GSK1070916 [104].

Adenosine is an another extracellular signaling molecule which is involved in numerous physiological functions throughout the brain and periphery and activates membrane-bound GPCRs [105] including adenosine receptors (A1AR, A2AAR, A2BAR, and A3AR) [[105], [106], [107], [108]]. A1AR in the brain slows metabolic activity for example at it reduces synaptic vesicle release at neuronal synapses and is implicated in sleep promotion [109], antiseizure as well as neuroprotective activity [108]. A1AR activation can also inhibit presynaptic Ca2+ influx-related release of transmitters to provide neuroprotection [110]. A3AR activation reduces inflammation and chronic neuropathic pain [107]. Selective compounds for the AR subtypes can potentially be used to control addictive psychostimulant consumption along with many other health problems [111]. We used machine learning models to find novel modulators with public data for A1AR agonists to generate and validate a Bayesian machine learning model and to identify molecules for testing [112]. Febuxostat and paroxetine were two FDA approved drugs which we identified as inhibitors of orthosteric radioligand binding for A2AAR and A3AR. For human A2AAR, stimulation of cAMP was observed for crisaborole (EC50 2.8 ​μM) and paroxetine (EC50 14 ​μM). Crisaborole also increased cAMP accumulation in A2BAR-expressing HEK293 ​cells. Paroxetine was found to bind human A3AR (Ki value of 14.5 ​μM), indicating antagonist activity [112]. These represent a few examples of where machine learning could be applied to GPCRs.

Kinase machine learning models

Glycogen Synthase Kinase 3 (GSK-3) is a serine-threonine kinase with broad specificity involved in inflammation, cell growth, cell differentiation, and energy metabolism [113,114]. It is also associated with age-related diseases like diabetes, AD, cancer [114,115] as well as rare diseases such Rett syndrome [116] and cyclin-dependent kinase-like 5 (CDKL5) disorder [117]. GSK-3 exists as two isoforms, GSK-3α and GSK-3β, with the latter found in higher abundance in the CNS and increases with age [118]. GSK-3β is also key in both peripheral and central inflammation, where it mediates inflammatory responses in astrocytes and microglia [[119], [120], [121]]. Inhibition of GSK-3β activates the Wnt pathway to promote neurogenesis [122] and therefore could be used to treat CNS diseases [123]. The brains of AD patients demonstrate increased levels of activated GSK3β [124,125], which phosphorylates the microtubule-stabilizing protein tau [[126], [127], [128]]. GSK3β inhibition also downregulates expression of soluble paired helical filaments, which are the major component of neurofibrillary tangles (NFTs) which are intracellular plaques [127,129,130]. GSK3β activity is further linked to the improper processing of the amyloid-precursor protein APP [131]. GSK3β inhibition downregulates expression of the β-site APP-cleaving enzyme 1 (BACE1) and the BACE1 gene reducing aβ production [132]. Tau hyperphosphorylation can hence be reduced by inhibiting GSK-3 [133] e.g. by PKC, which in turn is stimulated by M1 receptor agonists, including ACh, and a number of recent studies have proven that such products can reduce tau phosphorylation and aggregation [134]. While there are dozens of GSK3 inhibitors, most of these inhibitors are not suitable as therapeutics due to their toxicity [135], although some have displayed anti-AD activity in rodent models including lithium chloride [136], kenpaullones, tideglusib [137], 2-(alkylmorpholin-4-yl)-6-(3-fluoropyridin-4-yl)- pyrimidin-4(3H)-ones [138], isonicotinamides [139], pyrazine analogs [140], andrographolide [141], SAR502250 [142], AM404 [143], PF-367 [144]. Few GSK3β inhibitors have made it to clinical trials for AD or other diseases and none have shown promise in treating the disease [145,146]. We initially developed a GSK3β Bayesian machine learning model with 2368 molecules from ChEMBL (five-fold cross validation ROC of 0.905) which was used for virtual screening libraries of FDA approved drugs and clinical candidates [147]. In vitro testing of 5 selected compounds identified ruboxistaurin (GSK3β avg IC50 ​= ​97.3 ​nM) and 2 other diverse inhibitors as hits. Ruboxistaurin (LY-333531) is a protein kinase C (PKC) inhibitor. Our more recent machine learning studies have used 2618 GSK3β inhibitors to build regression models with 7 different algorithms with the SVR model performing the best with 5-fold cross validation (MAE 0.52) [148]. These models could be used to screen much larger datasets to identify novel scaffolds that could overcome limitations in the molecules that have reached the clinic.

Ion channel machine learning models

Voltage-gated sodium channels (VGSCs) such as Nav1.1, Nav1.6, Nav1.7, Nav1.8, and Nav1.9 which are expressed by adult sensory neurons [149] are known to be key determinants of sensory neuron excitability essential for: initial transduction of sensory stimuli, electrogenesis of the action potential, and neurotransmitter release. Therefore these ion channels are involved in chronic pain due to injury and disease caused by sensitization of the somatosensory nervous system involving these primary sensory neurons. Changes in expression of VGSCs, as well as post-translational modifications, can contribute to the sensitization of sensory neurons in these chronic pain states. Additionally, the gene variants of Nav1.7, Nav1.8, and Nav1.9 have been linked to human Mendelian pain disorders [150,151]. For example, Nav1.8 is a sodium ion channel voltage-gated channel subtype that is tetrodotoxin (TTX)-resistant and encoded by the SCN10A gene in humans [[152], [153], [154], [155]]. Nav1.8 has a role in nociception and is also expressed in the dorsal root ganglion (DRG), in unmyelinated, small-diameter sensory neurons called C-fibers [156,157]. As Nav1.8 is located in sensory neurons of the DRG this makes it a potential target for the development of new analgesics [158] for chronic pain [159]. Several drug companies have described Nav1.8 inhibitors [160] including the suzetrigine from Vertex [161,162] which was recently approved by the FDA. Nav1.8 was of initial interest to us as a validated target for the rare disease Pitt Hopkins Syndrome [163]. We initially screened the 1280 compound Prestwick chemical library using a Nav1.8 FLIPR high throughput screen and identified 93 hits (7.2 ​% hit rate) at >2SD from the mean response (Z’ ​= ​0.6). This screen also identified numerous dihydropyridine calcium channel inhibitors such as nicardipine which were further tested using automated patch clamping (nicardipine IC50 of 0.6 ​μM). At the same time, we used our Assay Central software to develop Bayesian machine learning models using ECFP6 descriptors in order to develop a model with this Nav1.8 inhibition data (5-fold cross validation showed an ROC of 0.84 [164]). Outside of this study we further curated literature data from ChEMBL on several additional sodium channels in order to build machine learning models (Nav 1.4 (ROC 0.80), Nav 1.5 (0.87) and Nav 1.7 (0.84)). In total, these models could also be used for virtual screening to identify potential selective inhibitors for each of these sodium ion channels.

Predicting molecule liabilities

Many drug candidate failures can be attributed to inadequate efficacy in animal models or in late stage clinical trials, indicating that improving the understanding of the candidates action in more complex systems could increase overall success [165]. In vitro or computational screening could be critical in doing this, while machine learning methods are being increasingly utilized there is still a shortage of datasets for in vivo endpoints. It is subsequently impossible to computationally determine these complex biological phenomena [166]. Increasingly, drug screening efforts skip to animal experiments which does not help when there is pressure to reduce animal use (e.g. more recently from the FDA and other regulators), and these efforts also require milligrams to grams of precious active compound to be synthesized, which may be far out of reach for those small companies that are resource constrained and likely working on rare and neglected diseases. It is at this juncture that computational approaches as an alternative may have their moment to impress. The following represent several examples of where computational approaches can be used to predict these molecule liabilities relevant for CNS drug discovery (Table 2).

Table 2.

Machine learning model statistics for CNS properties. B, Bayesian; BBB, blood brain barrier; DL, deep learning; RF, random forest; SVC, support vector classification, XGB, XGBoost.

Model N 5- fold AUC Model type External validation Reference
Hallucinogenic potential Psychlight B 84
PIKHAL and TIKHAL 221
0.76
0.67
SVC
B
22 molecules
22 molecules
[174]
[174]
Drug induced seizures 67 targets with different numbers of molecules – led to ∼2000 individual models - 43−5470 molecules per target representing 59,419 molecules Average 0.88 RF, SVC, XGBoost 25 ​% leave-out set, one hot encoding and ProtBERT were also used and did not improve predictions. [176]
BBB 2296 0.94 B Tested on 40 external molecules known to cross the BBB – 92.5 ​% correctly predicted. [200]

Predicting hallucinogenic potential

Psychiatric medicine is undergoing increased interest with psychedelics effectively treating numerous hard to treat diseases like substance abuse disorders, depression and previously treatment-resistant depression [[167], [168], [169]]. Psychedelics have a checkered history, are legally controlled in most countries and may act as agonists of serotonin 5-HT2A, as well as other receptors (e.g. dopaminergic and glutamatergic systems), to induce neuronal structure changes [169,170]. Development of psychoplastogens that avoid the psychedelic effects might benefit from efforts to model and predict this potential. Recent efforts to develop biosensor assays for 5-HT2A such as PsychLight [171] was used in vivo and in vitro. A human in vivo hallucinogenic dataset [172,173] covers phenyethylamines and tryptamines. These datasets were used with various machine learning approaches including pretrained large language models (MolBART), conformal predictors and with various molecular fingerprints to predict potential psychedelics [174]. Conformal predictors generally improved the predictive accuracy of our machine learning models over support vector classification (SVC) and removed test set predictions with high uncertainty. We also curated molecules that were tested in mouse head-twitch assays and a set of 5HT2A agonists [175] to test the models [174]. These models may offer an alternative approach to predict hallucinogenic potential prior to in vitro or in vivo testing.

Predicting Drug Induced Seizures

Drug Induced Seizures (DIS) are a component of CNS toxicity which leads to the high clinical failure rate observed of such drugs. Being able to predict this potential earlier in the drug discovery process would therefore enable us to likely avoid these failures and prioritize testing in experimental models of DIS. We have developed machine learning models for 67 target proteins implicated in seizures to predict this liability [176]. We also compiled a list of seizure liability compounds from various public references including a machine learning paper from 2011 that focused on predicting seizure liability (771 compounds) [177] as well as additional papers assessing the predictivity of their in vitro seizure liability models [[178], [179], [180], [181], [182], [183], [184]]. We also included the dataset (described earlier) which we compiled [185]. Once combined, this training dataset comprised 966 unique compounds (453 active, 543 inactive). Models were built using both ECFP6 and MACCS keys. Overall, these models had reasonable 5-fold cross validation scores with both ECFP6 descriptors (Fig. 3A) and MACCS keys (Fig. 3C), with the latter performing better overall. Example ROC plots are also shown for the best algorithms, specifically for K-nearest neighbors (knn) and random forest (rf) for ECFP6 and MACCS keys, respectively. These models represent a generalizable way to mitigate the potential of seizure liability early in the process in drug development.

Fig. 3.

Fig. 3

Summary of the 5-fold cross validation metrics of models predicting general seizure liability using either (A,B) ECFP6 or (C,D) MACCS keys molecular descriptors. ROC curves are also shown for the best performing algorithms K- nearest neighbors (knn) (B) and random forest (rf) (D).

Blood brain barrier models

Considering the efficiency of the BBB, it is difficult to design pharmaceutical products that can effectively be delivered to the CNS. The BBB changes in response to each pathological condition, requiring extensive knowledge of how to protect the CNS from possible challenges or inflammatory immune cells while providing the necessary therapeutics. Various biological, chemical, and physical methods exist to open the BBB, including the use of cyclodextrin, viruses, and microwaves [186]. Some approaches for traversing the BBB include opening tight junctions, transendothelial passage, efflux pump disruption, and targeted receptor mediated transport. Our first published efforts modeling BBB were from 2008 [187] when we and others had published coincidently and reached the conclusion that such models were probably at their peak [187]. While available datasets are now many times larger, their relevance to the human BBB is unchanged. There are now many machine learning models or rules developed to predict the ideal molecular properties to cross the BBB in mouse [188,189] as well as quantitative structure property relationships (QSPR) [190] and various types of machine learning models [[191], [192], [193], [194], [195], [196], [197]]. Wager et al. published their analysis of 119 CNS drugs and developed a method to score 108 CNS candidates from Pfizer programs with a multiparameter optimization (MPO) algorithm [198] using molecular properties (ClogP, ClogD, MW, TPSA, HBD and pKa). This correctly scored 74 ​% of a set of 119 marketed drugs while 60 ​% of Pfizer's own drug candidates scored favorably with this MPO score. Another example tested an updated method on 21 CNS candidates with 77 ​% correct [199]. We recently reimplemented these methods [200] using ChemAxon descriptors to replace the ACD software used originally [198] as well as developed a Bayesian BBB machine learning model based on published data for over 2000 molecules. Evaluating the later model on 40 molecules from the MPO dataset demonstrated >90 ​% correct predictions and illustrated that such methods could have benefits over relatively simple rules like the MPO method [200]. These BBB models will likely not be perfect, as we have seen over the decades there are issues around the datasets used and the general lack of prospective validation. We have also seen pharmaceutical companies share results, but they generally do not share the underlying mouse BBB data relating to proprietary compounds.

AI and Machine Learning approaches for drug discovery of neurological disorders

The impact of AI and machine learning on drug discovery in the areas of neurodevelopmental disorders, depression, PD, AD, anesthesia and pain treatment has been reviewed, providing select examples [201]. There are numerous examples of the use of different AI and machine learning approaches integrating different types of data at various stages of the drug discovery continuum and what follows is only a small sampling in the interests of space. It should be noted that most of the CNS-related multimodal approaches are for clinical applications (MRI, CT data etc.) rather than for drug discovery. What follows are various applications of these AI and machine learning approaches applied to various neurological disorders.

Knowledge graphs and their applications

A large knowledge graph was described which used data from 29 public sources that captured data on 1552 small molecules and 137 complex diseases [202]. A machine learning approach then used this data to predict the probability of treatment (AUC ROC ​= ​97.4 ​%), leading to good predictions for DrugCentral and novel clinical trial indications resulted. Two case studies were provided, one being nicotine dependence and this provided molecules already used or in clinical trials for smoking cessation (e.g. galantamine). A second case study was epilepsy, and this provided some known anti-epileptics as well as novel compounds such as acamprosate (a GABA-A receptor modulator). In addition, 15 additional molecules that cause epilepsy were also provided [202]. The results of this approach would have been more impressive if prospective testing had been performed.

IBM Watson for Drug Discovery was used to build a knowledge network between psychiatric and neurological disorders, 1588 genes and 722 drugs from scientific publications over the past 50 years [203]. The network had 2 distinct groups including psychiatric diseases (anxiety, depression, schizophrenia, autism and bipolar disorder) and neurodegenerative diseases (AD, PD, HD and dementia). In the process, drugs targeting genes related to these diseases were uncovered that have yet to be used to treat them, including monoclonal antibodies (anti-CD33). The approach can also be validated by identifying drugs that are in clinical studies for treating the same diseases, but which lacked publications [203]. Again, a limitation of this study is that they did not however test any compounds to validate the predictions.

Using genome wide association studies, a network topology based deep learning framework NETTAG was used to identify AD disease associated genes [204]. Using this approach 156 AD-risk genes were found to be enriched in druggable targets. Network proximity approaches were used to computationally identify 118 high scoring candidates from nearly 3000 FDA approved drugs and gemfibrozil (a lipid regulator which is used in hypercholesterolemia) was top scoring. Using a database to search medical data on over 10 million patients, gemfibrozil was also identified as associated with decreased AD incidence as patients showed a 43 ​% decrease in disease risk [204].

Natural language processing has been used for mining over 30 million articles in PubMed to identify text relationships between diseases, genes and proteins to construct a knowledge graph using SemNet 2.0 and rank molecules [205]. This was then used to aid repurposing of drugs for PD starting from the known drug levodopa. Several antihistamines were identified (including ebastine, levocetirizine) the antibiotic vancomycin, the NMDA agonist neramexane and the angiotensin converting enzyme (ACE) inhibitor captopril. In several of these cases e.g. antihistamines, there was good evidence in animal models. These repurposed drugs have a role in oxidative stress, neurotransmitter imbalance, and inflammation. Limitations of the approach are the proposal of drugs that may not be available or approved in the US as well as having possible undesirable side effects. In addition, crossing the BBB is also important and some of those selected may not be able to [205].

Generative machine learning and transformer approaches

The evolution over 50 years from quantitative structure activity relationships (QSAR) to machine learning for neurological drug discovery has been briefly summarized [206]. It has evolved from limited data and small regression models to comparably very large datasets. Several examples were provided of molecules like the BACE-1 inhibitor elenbecestat, which was identified by machine learning, that ultimately failed in a phase III trial for AD. A second example was the AI designed molecule DSP-1181 for obsessive compulsive disorder which reached the clinic in 2019. It was suggested that the future of machine learning in neurodegenerative drug discovery is likely linked to integration with other approaches like personalized medicine and imaging [206].

Most of the machine learning examples described thus far use a model to score an array of molecules to predict a bioactivity or a property (e.g. BBB). Ideally there are multiple properties alongside bioactivity that need optimization, such that we could also start with a desired property value and design molecules that fulfill these requirements. A generative model is one way to achieve this as it learns the probability of a chemical with the desired properties given the structure [207]. There have been numerous discoveries in the field of de novo drug design [208,209] and several different algorithms such as Generative Adversarial Networks (GANs) [210], Variational Autoencoder (VAE) [211,212] and Recurrent Neural Networks (RNN) [209] have been used to design molecules for various targets [[213], [214], [215], [216]]. These methods can use different molecular representations including 2D strings [217,218] or 3D graphs [219]. Increasingly, examples include training with molecules from ChEMBL or other large sources of molecules (e.g. USPTO), using such methods as a two-layer long short term memory (LSTM)-based RNN that was used to generate libraries of molecules as SMILES (one dimensional strings) followed by transfer learning to tune the model and then generate a library of leads from a starting known active fragment [220]. Another example generated molecules based on predicted molecular properties [221]. Most of the published generative examples have optimized against a single physicochemical activity or a bioactivity and clearly this is a limitation as this will not help if the molecule needs selectivity against closely related targets or to avoid off-target effects. Several groups have also focused on graph structures which had returned 100 ​% valid structures [219]. Other algorithms such as transformers and attention-based models have also been used. The biggest challenge for these generative approaches is the limited prospective experimental validation which involves synthesizing compounds and testing for activity [222] or finding structurally similar compounds to those designed from vendors [223]. Generative models can propose molecules like a starting structure, explore chemistry space or fit design criteria such that a single generative model may not be able to respond to all the desired molecule design needs.

De novo design approaches generally have been frequently used to design molecules for a single target, however, there have also been efforts to also do this for multiple targets simultaneously. One example is MTMol-GPT a multitarget generative pretrained transformer model which uses a dual discriminator model with inverse reinforcement learning [224]. Both the 5HT1A and dopamine receptor D2 were targeted for neuropsychiatric applications. Docking (AutoDock Vina) and pharmacophore mapping were then used to evaluate 1000 designed ligands. In general, the molecules had improved docking scores compared to known ligands. There was also good mapping to pharmacophore features. It should be noted that none of these molecules were synthesized and tested, while there was also no comparison of how far the molecules are from known ligands for both receptors [224].

Neuropeptides are produced by neurons attached to GPCRs and they play important roles in modulation of brain function including sleep, food intake and have neuroprotective roles that may be important for PD and AD. A recent machine learning model trained on 6807 neuropeptides (5–100 amino acid residues) and 8647 non-neuropeptides collected from public databases used a two-phase approach in which a bidirectional encoder representation from transformers (BERT), capturing the functional and structural aspects of the neuropeptide then a non-dominated sorting genetic algorithm which explores and changes the population of neuropeptides [225]. 80 ​% of the data was used for training while 10 ​% was used for validation and a further 10 ​% for testing. The BERT-NeuroPred model had a 91 ​% accuracy, however again no external testing with prospective designs was performed. Secondary and tertiary characteristics of neuropeptides were also not incorporated into the objectives of the model [225].

Docking and other applications with machine learning

Docking of small molecules could likely populate many reviews on the topic. Challenges here include the scoring function used to rank molecules before selection. Applications of docking for neurological diseases combined with machine learning reveals few examples. For example, α-synuclein aggregation is linked to the neurodegeneration which occurs in PD, therefore drugs that can block this can be disease modifying [226]. Docking of 2 million molecules to α-synuclein fibrils led to 79 compounds of which 68 were tested and 4 were found to be active. Similarity searching resulted in additional hits and enabled a training set for machine learning of 161 molecules. These models iteratively enabled the selection of more molecules for testing and showed 20-fold higher hit rates than high throughput screening as well as increased novelty. Limitations of this approach included screening a library of existing molecules as well as focusing on just one parameter [226].

As the human interactome is massive with ∼650,000 protein-protein interactions (PPI) these have key roles that if dysregulated they can cause disease and hence represent drug targets. There are therefore few if any examples of PPI inhibitors developed by AI specifically for CNS targets. A recent review summarized the evolution of computational approaches in this area and was largely silent on CNS targets [227] so there is certainly a potential opportunity here.

Replacing animal models with AI

In the area of AI in neuroregeneration, only 12 ​% of the papers relate to drug discovery, with most relating to diagnosis of disease and rehabilitation, and the methods were used for analysis of imaging data as well as animal models [228]. Much of neurological research uses animal experiments, but new AI approaches may offer one way to replace these such that the models are more human centric. The scope of such applications can reduce the need for animals in screening, length of longitudinal studies to understand disease progression, diagnostic development, neurotoxicity testing, minimize studies for brain computer interface development and potentially replace some developmental testing. By accumulating published examples that illustrated this potential of AI in neurology research it set the foundation for exploring how AI might evolve in future and the potential for impact in different areas [229]. Some limitations were noted such as relying on data from certain populations for model building which could bias models, overfitting of data due to misleading predictions from messy data, and predictions that may be difficult to explain. While very futuristic this review made the point that AI has the potential to replace animal experiments in neurology [229]. One might argue also that this might be feasible in other disease areas too.

Behavioral screening for discovery of neuroactive drugs does not have to rely on rodents or other animals, such as the even the aquatic invertebrate the freshwater planarian, to generate phenotypic data [230]. This can be used alongside physicochemical property data of molecules to generate classifiers for prediction of phenotype. One proof of concept study used 19 neuroactive molecules spanning anxiolytics, antidepressants and antipsychotics. The classification accuracy using PCA and other projection methods was similar for physicochemical and phenotypic data [230]. While this was a very small dataset there was no cross validation or external predictions, it does leave out the possibility that a larger training set would be of more potential value. Insects such as locust have also been used to model brain uptake of drugs. A recent study generated data on 25 drugs and used this to build machine learning models with simple interpretable descriptors to predict brain uptake [231]. The small dataset was used with cross validation and a hold-out test set to produce acceptable results, again suggesting the potential of this system for screening compounds and building models with bigger datasets. As yet these few examples suggest too little data for building machine learning models that could be used for general screening for this ability to reach the brain.

Clinical data applications of AI

Compared to using AI in drug discovery, applications to clinical data are relatively rare. One study integrated clinical whole genome sequencing and genetic data from 235 patients to build various machine learning models with nested cross validation to predict the response for the antiepileptic brivaracetam [232]. 106 clinical and 40 genetic features (4695 genetic variants) were used to evaluate different model types. A gradient-boosted tree model performed the best on cross validation (AUC 0.76) and with an external test set of 47 patient samples (AUC 0.75) to validate the model. Interestingly the same study also assessed the placebo response using the clinical features only, but was unable to predict this response better than random [232].

Approaches identifying potential clinical candidates for diseases include searching for associations across medical records for large numbers of patients using machine learning approaches. When many of these studies are performed globally, they can be combined into a systematic review. Efforts to do this to find drugs that modify dementia risk or AD yielded several different drug classes such as vaccinations, antimicrobials and anti-inflammatories that decrease risk while diabetes drugs and antipsychotics increased risk. Some classes such as antidepressants and antihypertensives produce conflicting evidence [233]. Again, this type of approach could be readily applied to other neurological diseases outside of those related to dementia.

The application of AI to dementia drug discovery and clinical trial design was recently reviewed and one example focused on unsupervised approaches to identify subgroups or progression patterns within cross-sectional databases, while another approach described subtyping AD using machine learning from RNA data [234].

Continuous electroencephalography (cEEG) is used for detection of seizures as well as monitoring depth of sedation and ischemia. Increased use of the technique has also led to the detection of non-convulsive seizures [235]. Challenges with the approach include the review of the data. AI has been applied to seizure prediction using machine learning to produce risk scores, EEG screening, automated annotation and interpretation of the EEG, seizure management, the diagnosis of non-convulsive seizures and paroxysmal events, treatment of seizures and status epilepticus, detection of cerebral ischemia in subarachnoid hemorrhage, neuroprognostication in traumatic and anoxic brain injury, as well as anesthetic dosing and sedation management. Even in this area, there are barriers including computing resources, lack of prospective studies, generalizability of approaches due to biases in training data, the need for larger training sets as well as lack of validation that need to be considered [235].

Drug repurposing in AD (DRIAD) is a machine learning approach which quantifies associations in AD pathology severity and molecular mechanisms [236]. When combined with human neural cell cultures exposed to 80 FDA approved predominantly kinase inhibitor drugs and RNA-seq performed resulting in gene lists that can be ranked to prioritize the compounds. The JAK inhibitor ruxolitinib scored well and suggested the JAK-STAT pathway is important in AD and worth further exploration. The results of the study were not further validated, but this could be partially aided by searching electronic health records. The BBB penetration of the compounds was also not evaluated so it is also possible while the compounds may look promising in neural cells in vitro, the delivery in vivo may be challenging and limit the value [236].

Phenotypic screening and imaging applications with AI

One early example of predicting neuropharmacological potential uses a mouse screening system that leverages computer vision to track their behavior and extract different measures for an array of 14 different compound classes called SmartCube [237]. This enabled the creation of signatures for each drug class and prospective prediction was demonstrated with 2 compounds (TP-10 and PF-670462) not in the model, one was predicted to be anxiolytic and the other an antipsychotic. These predictions were followed up with further animal studies to confirm the behavior predicted from the screen [237].

Live cell imaging of neuronal cells and screening of molecules is important for identifying new therapeutics. Screening of neuronal cell cultures combined with machine learning offers a powerful approach for learning from the imaging data to identify patterns that may be missed by human observers [238]. The development of automated imaging systems to generate large-standardized datasets for such models will also be important.

Algorithms that work well with small datasets include few-shot learning. Few shot meta learning allows models to learn from prior experiences and improve the learning performance. By combining this approach with a high throughput automated whole brain activity mapping (BAM) screen in zebrafish it is possible to improve the prediction of molecules for CNS agents like those for PD [239].

Neuroactive drugs can be repurposed for other uses. For example, a set of 132 such neuroactive drugs were profiled in glioblastoma patient material from 27 patients using single cell transcriptomes [240]. The convergence of secondary drug targets was analyzed by regularized regression (COSTAR). Logistic LASSO regression has been used to train a model to identify the secondary target genes that discriminated hit drugs from non-hits to derive a signature predictive of the ex-vivo efficacy for glioblastoma. A further 1.1 million molecules were screened with this signature model of which 23 top scoring and 25 bottom scoring molecules were tested across 4 patient samples (predictions and scores AUC 0.94) and only the hits were linked to the target BTG [240].

Reinforcement learning (RL) can be used to tune a generative model for the properties of interest and deliver molecules with a different property distribution when compared to those of a training set [241]. This method is however inefficient when compared to other methods and will take a long time to train, hence such methods are usually combined with adversarial or pretraining. RL can bridge target-based and phenotype-based drug discovery. A recent review has provided an overview of RL and may be used for systems-pharmacology in neurological disorders as well as other applications [241]. As yet these various imaging application of machine learning are in their nascent stages.

Designing illicit substances targeting the CNS using AI

A further unusual application of computational approaches is in trying to get ahead of the problem of illicit substances and one such area is in novel psychoactive substances (NPS) which are a global issue and are generally heavily regulated [242]. However, there is a consistent approach from violators where they subtly modify the molecules to circumvent legislation. A scaffold and transformer based NPS generation and screening (STNS) has been used for chemical space analysis. A discriminator model was generated from 2154 NPS molecules and 2154 negative samples from PubChem. The NPS generator is a generative model composed of T-Scaffold and Mol-GPT which was used to design over 120,000 unique cannabinoid molecules. The model was then validated and 3 novel, synthetic cannabinoids were identified that docked well in the cannabinoid receptor and were tested in vitro with a genetically encoded endocannabinoid (eCB) sensor (GRABeCB2.5) [242]. While not addressed in the article, the likely dual use nature of the work is clearly apparent as is the potential to repurpose some of these illicit molecules as potential therapeutics for neurological diseases. To our knowledge we have not seen examples in the literature, were machine learning has been used by criminals to create illicit CNS targeted molecules that have then been distributed and used. We have highlighted this potential capability in the area of nerve agents [243] and the software and datasets are clearly available.

A generative approach to developing treatments for opioid use disorder

Opioid abuse leads to complex and profound changes in the brain. Opioid use disorder (OUD) induces changes in the mesolimbic reward circuit, and these changes are thought to underlie the pathology and cycles of addiction [244]. Abrupt disuse of opioids leads to opiate withdrawal, with common symptoms including aches and pains, muscle spasms, rapid sensation of temperature swings, tremors, nausea and vomiting, anxiety and irritability, and insomnia [245]. Often times, despite the initial reasoning for opioid use, OUD occurs due to the desire to avoid these withdrawal symptoms [246]. The 5-HT2R family members, namely 5-HT2A and 5-HT2C, are directly involved in mediating neurotransmission in substance use disorder [247]. 5-HT2A and 5-HT2C are widely expressed in the brain and bring about the abuse-related effects of drugs such as cocaine, morphine and heroin through their interactions with the dopamine system (DA) [[248], [249], [250], [251], [252], [253], [254]]. Recently, a new class of molecules derived from psychedelics have been introduced known as psychoplastogens [255,256]. These molecules are described as various serotonergic and entactogens that rapidly display plasticity-promoting properties by activation of mTOR. The ability to induce neuroplasticity is thought to be the end result of increased BDNF release, which stimulates spinogenesis and neuritogenesis [257]. Critically, the psychedelic experience and induced neuronal plasticity are not entirely linked, and some evidence suggests that the psychedelic experience is separate from the therapeutic role of psychedelics [258]. While the 5-HT2A receptor is critical for both the psychedelic experience and neuroplasticity, some psychoplastogens do not induce a psychedelic experience [256]. Recently, an ibogaine analog was synthesized which decreased drug-seeking responses and induced neuritogenesis and spinogenesis in rats while not inducing a head-twitch response, a common hallmark of a psychedelic experience in rodents [256]. This ibogaine analog was specific to 5-HT2A, unlike traditional psychedelics which broadly target other 5-HT receptors as well. Psychedelic response has been attributed to the activation of 5-HT2A receptors [259], and 5HT2C has been targeted by molecules as potential treatments for addiction [[260], [261], [262], [263], [264], [265], [266]], suggesting that an appropriately selective serotonin agonist for either could treat OUD, although to date clinical translation has been mixed likely due to the small studies [[267], [268], [269], [270]].

We have recently described our generative drug discovery software MegaSyn and applied it to generating ibogaine analogs [271] that could be used for OUD. This model was first trained on a subset of ChEMBL representing ∼2 million compounds. Next, we applied transfer-learning by training for an epoch on a natural products library [272] then refined the RNN using a hill-climb maximum-likelihood estimator algorithm. The top 10 ​% of ranked compounds were kept and fed back into the model for training. The model starts by generating drug-like compounds is re-trained on the top scoring compounds until it ultimately finds those molecules that have the desired chemical properties. As a scoring reward function the Tanimoto similarity to the CANVASS library [272] (>0.2) was used as well as an activity model against 5-HT2A (predicted active), and avoiding off-target models (hERG, 5-HT1A, 5-HT1F, 5-HT2C). We wanted to obtain molecules with Tanimoto similarity >0.6 to Ibogaine and a lower cLogP. After generating 100,000 virtual molecules, all of the top 50 generated molecules had higher scores versus ibogaine and we were able to demonstrate that MegaSyn produced the psychoplastogen tabernanthalog [256] suggesting that it could discover molecules with the properties desired in this case [271]. We have recently further updated this approach (Fig. 4) and used MegaSyn in house for the design of additional molecules targeting serotonin receptors with good BBB penetration as well as other receptors [271,273] which have been validated after synthesis and testing in vitro.

Fig. 4.

Fig. 4

A schematic overview of the MegaSyn generative approach to molecule optimization training, which consists of nested training loops [271,273]. Targets A-C could be individual CNS receptors of interest for which one desires selectivity, or they could be multiple targets that need to be hit to obtain polypharmacology goals.

Summary, limitations and future perspective

As neurological disease encompasses over 1000 disorders it would be impossible to cover them all in this review! Clearly only a fraction of these diseases has been the focus of machine learning and AI to date, in general those with larger populations and for which there is more published data. The costs of research and development require a rethink in how we can address developing treatments for more diseases which impact human health and have a dramatic financial toll particularly those targeting the CNS. Most of the diseases that have achieved the greatest attention from these efforts are those neurological diseases that are highly complex and heterogeneous which also affect the largest numbers of patients. Yet the diseases that are likely to be the most straightforward which are monogenic (and rare) have not received much attention from such machine learning and AI approaches [[274], [275], [276]]. There are few if any drugs for neurological diseases that are derived from machine learning or AI yet, although that may change soon. It is important to note that these computational approaches have been developing for many decades and are only now achieving a high level of recognition likely due to increased attention. This may also be due to the intersection of availability of large amounts of public data such as structure activity data (e.g. ChEMBL, BindingDB etc.), genomics, proteomics, metabolomics, increased computational power available (e.g. GPUs), more advanced algorithms (transformers, deep learning etc.), etc.

Just having the data and the software/hardware tools available though does not mean that the models will be built or used for drug discovery. There must be the motivation, the funding and know how to do this work. Someone has to do it! As we have described, much of the computational work is done entirely with little to no experimental validation. This is a substantial limitation. In the published examples there is a clear bias towards success, failed efforts are not discussed and limitations in the underlying data or approaches are rarely considered by the authors. If this is to change in future to impact neurological disease research, we need to be training more multidisciplinary scientists who can handle not only the computational side but also the experimental piece as well. Certainly, collaborations between computational and experimental scientists could also achieve the same end but these will need to happen and bringing together the right team is as much an art as science.

Discovering drugs is extremely difficult and perhaps more so for neurological diseases where you need more data such as BBB permeability and an understanding of any off-target effects with an array of GPCRs, enzymes or ion channels where signal dysregulation could lead to hallucinogenic effects or seizures. An added part of the challenge is not just computational, but the animal models for these diseases poorly reflect the human neurological diseases, so it is more difficult to validate computational and in vitro findings prior to going into clinical trials in humans. We are clearly embarking on the age of AI for neurological diseases with great promise although it is unclear whether it will have any more impact than any other technology in drug discovery that has been heralded as transformational over the last 30 years. This is another significant question that remains unanswered. We have described some of the nascent machine learning and AI applications whether they are individual techniques or consist of end-to end applications. Some of the more basic applications we have predominantly used and highlighted, help to focus the universe of molecules for testing in a single assay whereas some of the more complex generative approaches have the promise to design molecules with ideal properties such that few must be made or tested against one or more targets or the disease of interest. But again, we need more examples to convince other scientists and the industry in general that this is the future. We hope this review serves to inspire others to consider applying the currently commercial or academic available machine learning and AI tools to their neurological disease of interest. Alternatively, there are likely also many new opportunities to develop completely new software or applications to make the discoveries necessary for identification of new treatments for neurological diseases where there is still considerable unmet need. There are millions of patients globally with neurological diseases whether common or rare, and they need treatments urgently. That should provide the motivation to use and improve the tools we have described in order to find these transformational drugs.

Funding

We kindly acknowledge NIH funding from R44GM122196-02A1 from NIGMS and 1R44ES031038-01 from NIEHS for our machine learning software development and applications. “Research reported in this publication was supported by the National Institute of Environmental Health Sciences of the National Institutes of Health under Award Number R44ES031038. We also acknowledge 1R43DA055419-01 from NIDA. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.” NINDS have supported our work on Batten disease (2R44NS107079-02A1, 1SB1NS135733-01 from NIH/NINDS) which is briefly mentioned herein.

Declaration of competing interest

The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Sean Ekins reports financial support was provided by NIH National Institute of General Medical Sciences. Sean Ekins reports financial support was provided by National Institute of Environmental Health Sciences. Sean Ekins reports financial support was provided by National Institute on Drug Abuse. Sean Ekins reports a relationship with Collaborations Pharmaceuticals, Inc. that includes: employment. Sean Ekins has patent pending to Collaborations Pharmaceuticals, Inc. None If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

Dr's. Patricia A. Vignaux, Joshua Harris, Thane Jones, Renuka Raman, Ana C. Puhl, Fabio Urbina, Mustafa Kemal Ozalp, Kenneth Jacobson, Keith Pennypacker, Daniela Brunner and our colleagues are thanked for their contributions, discussions and collaborations. Dr. Barbara Slusher is kindly acknowledged for this opportunity to review the topic.

References

  • 1.Paul S.M., Mytelka D.S., Dunwiddie C.T., Persinger C.C., Munos B.H., Lindborg S.R., et al. How to improve R&D productivity: the pharmaceutical industry's grand challenge. Nat Rev Drug Discov. 2010;9(3):203–214. doi: 10.1038/nrd3078. [DOI] [PubMed] [Google Scholar]
  • 2.Wouters O.J., McKee M., Luyten J. Estimated research and development investment needed to bring a new medicine to market, 2009-2018. JAMA. 2020;323(9):844–853. doi: 10.1001/jama.2020.1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Galkina Cleary E., Jackson M.J., Zhou E.W., Ledley F.D. Comparison of research spending on new drug approvals by the national Institutes of health vs the pharmaceutical industry, 2010-2019. JAMA Health Forum. 2023;4(4) doi: 10.1001/jamahealthforum.2023.0511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Henderson R.H., French D., Stewart E., Smart D., Idica A., Redmond S., et al. Delivering the precision oncology paradigm: reduced R&D costs and greater return on investment through a companion diagnostic informed precision oncology medicines approach. J Pharm Policy Pract. 2023;16(1):84. doi: 10.1186/s40545-023-00590-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Schuhmacher A., Hinder M., Brief E., Gassmann O., Hartl D. Benchmarking R&D success rates of leading pharmaceutical companies: an empirical analysis of FDA approvals (2006–2022) Drug Discov Today. 2025;30(2) doi: 10.1016/j.drudis.2025.104291. [DOI] [PubMed] [Google Scholar]
  • 6.Pangalos M.N., Schechter L.E., Hurko O. Drug development for CNS disorders: strategies for balancing risk and reducing attrition. Nat Rev Drug Discov. 2007;6(7):521–532. doi: 10.1038/nrd2094. [DOI] [PubMed] [Google Scholar]
  • 7.Nelvagal H.R., Eaton S.L., Wang S.H., Eultgen E.M., Takahashi K., Le S.Q., et al. Cross-species efficacy of enzyme replacement therapy for CLN1 disease in mice and sheep. J Clin Investig. 2022;132(20) doi: 10.1172/JCI163107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ekins S., Puhl A.C., Zorn K.M., Lane T.R., Russo D.P., Klein J.J., et al. Exploiting machine learning for end-to-end drug discovery and development. Nat Mater. 2019;18(5):435–441. doi: 10.1038/s41563-019-0338-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mielke M.M., Vemuri P., Rocca W.A. Clinical epidemiology of Alzheimer's disease: assessing sex and gender differences. Clin Epidemiol. 2014;6:37–48. doi: 10.2147/CLEP.S37929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lee J.E., Han P.L. An update of animal models of Alzheimer disease with a reevaluation of plaque depositions. Exp Neurobiol. 2013;22(2):84–95. doi: 10.5607/en.2013.22.2.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.West S., Bhugra P. Emerging drug targets for Abeta and tau in Alzheimer's disease: a systematic review. Br J Clin Pharmacol. 2015;80(2):221–234. doi: 10.1111/bcp.12621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Devinsky O., Vezzani A., O'Brien T.J., Jette N., Scheffer I.E., de Curtis M., et al. Epilepsy. Nat Rev Dis Primers. 2018;4 doi: 10.1038/nrdp.2018.24. [DOI] [PubMed] [Google Scholar]
  • 13.Disease G.B.D., Injury I., Prevalence C. Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990-2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet. 2016;388(10053):1545–1602. doi: 10.1016/S0140-6736(16)31678-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cook D., Brown D., Alexander R., March R., Morgan P., Satterthwaite G., et al. Lessons learned from the fate of AstraZeneca's drug pipeline: a five-dimensional framework. Nat Rev Drug Discov. 2014;13(6):419–431. doi: 10.1038/nrd4309. [DOI] [PubMed] [Google Scholar]
  • 15.Roberts R., Authier S., Mellon R.D., Morton M., Suzuki I., Tjalkens R.B., et al. Can we panelize seizure? Toxicol Sci. 2021;179(1):3–13. doi: 10.1093/toxsci/kfaa167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Authier S., Arezzo J., Delatte M.S., Kallman M.J., Markgraf C., Paquette D., et al. Safety pharmacology investigations on the nervous system: an industry survey. J Pharmacol Toxicol Methods. 2016;81:37–46. doi: 10.1016/j.vascn.2016.06.001. [DOI] [PubMed] [Google Scholar]
  • 17.NIDA Drug overdose death rates. 2024. https://nida.nih.gov/research-topics/trends-statistics/overdose-death-rates
  • 18.Ropero-Miller J.D., Speaker P.J. The hidden costs of the opioid crisis and the implications for financial management in the public sector. Forensic Sci Int. 2019;1:227–238. doi: 10.1016/j.fsisyn.2019.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gooch C.L., Pracht E., Borenstein A.R. The burden of neurological disease in the United States: a summary report and call to action. Ann Neurol. 2017;81(4):479–484. doi: 10.1002/ana.24897. [DOI] [PubMed] [Google Scholar]
  • 20.Ruddigkeit L., Blum L.C., Reymond J.L. Visualization and virtual screening of the chemical universe database GDB-17. J Chem Inf Model. 2013;53(1):56–65. doi: 10.1021/ci300535x. [DOI] [PubMed] [Google Scholar]
  • 21.Jumper J., Evans R., Pritzel A., Green T., Figurnov M., Ronneberger O., et al. Highly accurate protein structure prediction with AlphaFold. Nature. 2021;596(7873):583–589. doi: 10.1038/s41586-021-03819-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Diaz-Holguin A., Saarinen M., Vo D.D., Sturchio A., Branzell N., Cabeza de Vaca I., et al. AlphaFold accelerated discovery of psychotropic agonists targeting the trace amine-associated receptor 1. Sci Adv. 2024;10(32) doi: 10.1126/sciadv.adn1524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ferreira-Vieira T.H., Guimaraes I.M., Silva F.R., Ribeiro F.M. Alzheimer's disease: targeting the cholinergic system. Curr Neuropharmacol. 2016;14(1):101–115. doi: 10.2174/1570159X13666150716165726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Komloova M., Musilek K., Dolezal M., Gunn-Moore F., Kuca K. Structure-activity relationship of quaternary acetylcholinesterase inhibitors - outlook for early myasthenia gravis treatment. Curr Med Chem. 2010;17(17):1810–1824. doi: 10.2174/092986710791111198. [DOI] [PubMed] [Google Scholar]
  • 25.Gonzalez J., Ramirez J., Schwans J.P. Evaluating Fmoc-amino acids as selective inhibitors of butyrylcholinesterase. Amino Acids. 2016;48(12):2755–2763. doi: 10.1007/s00726-016-2310-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Castro A., Martinez A. Peripheral and dual binding site acetylcholinesterase inhibitors: implications in treatment of Alzheimer's disease. Mini Rev Med Chem. 2001;1(3):267–272. doi: 10.2174/1389557013406864. [DOI] [PubMed] [Google Scholar]
  • 27.Kaur J., Zhang M.Q. Molecular modelling and QSAR of reversible acetylcholines-terase inhibitors. Curr Med Chem. 2000;7(3):273–294. doi: 10.2174/0929867003375254. [DOI] [PubMed] [Google Scholar]
  • 28.Tong W., Collantes E.R., Chen Y., Welsh W.J. A comparative molecular field analysis study of N-benzylpiperidines as acetylcholinesterase inhibitors. J Med Chem. 1996;39(2):380–387. doi: 10.1021/jm950704x. [DOI] [PubMed] [Google Scholar]
  • 29.Golbraikh A., Bernard P., Chretien J.R. Validation of protein-based alignment in 3D quantitative structure-activity relationships with CoMFA models. Eur J Med Chem. 2000;35(1):123–136. doi: 10.1016/s0223-5234(00)00108-2. [DOI] [PubMed] [Google Scholar]
  • 30.El Yazal J., Rao S.N., Mehl A., Slikker W., Jr. Prediction of organophosphorus acetylcholinesterase inhibition using three-dimensional quantitative structure-activity relationship (3D-QSAR) methods. Toxicol Sci. 2001;63(2):223–232. doi: 10.1093/toxsci/63.2.223. [DOI] [PubMed] [Google Scholar]
  • 31.Sutherland J.J., O'Brien L.A., Weaver D.F. A comparison of methods for modeling quantitative structure-activity relationships. J Med Chem. 2004;47(22):5541–5554. doi: 10.1021/jm0497141. [DOI] [PubMed] [Google Scholar]
  • 32.Fernandez M., Caballero J. Ensembles of Bayesian-regularized genetic neural networks for modeling of acetylcholinesterase inhibition by huprines. Chem Biol Drug Des. 2006;68(4):201–212. doi: 10.1111/j.1747-0285.2006.00435.x. [DOI] [PubMed] [Google Scholar]
  • 33.Jung M., Tak J., Lee Y., Jung Y. Quantitative structure-activity relationship (QSAR) of tacrine derivatives against acetylcholinesterase (AChE) activity using variable selections. Bioorg Med Chem Lett. 2007;17(4):1082–1090. doi: 10.1016/j.bmcl.2006.11.022. [DOI] [PubMed] [Google Scholar]
  • 34.Manchester J., Czermiński R. SAMFA: simplifying molecular descriptors for 3D-QSAR. J Chem Inf Model. 2008;48:1167–1173. doi: 10.1021/ci800009u. [DOI] [PubMed] [Google Scholar]
  • 35.Akula N., Lecanu L., Greeson J., Papadopoulos V. 3D QSAR studies of AChE inhibitors based on molecular docking scores and CoMFA. Bioorg Med Chem Lett. 2006;16(24):6277–6280. doi: 10.1016/j.bmcl.2006.09.030. [DOI] [PubMed] [Google Scholar]
  • 36.Shiri F., Pirhadi S., Ghasemi J.B. Dynamic structure based pharmacophore modeling of the Acetylcholinesterase reveals several potential inhibitors. J Biomol Struct Dyn. 2019;37(7):1800–1812. doi: 10.1080/07391102.2018.1468281. [DOI] [PubMed] [Google Scholar]
  • 37.Chekmarev D., Kholodovych V., Kortagere S., Welsh W.J., Ekins S. Predicting inhibitors of acetylcholinesterase by regression and classification machine learning approaches with combinations of molecular descriptors. Pharm Res. 2009;26(9):2216–2224. doi: 10.1007/s11095-009-9937-8. [DOI] [PubMed] [Google Scholar]
  • 38.Zhang L., Li D., Cao F., Xiao W., Zhao L., Ding G., et al. Identification of human acetylcholinesterase inhibitors from the constituents of EGb761 by modeling docking and molecular dynamics simulations. Comb Chem High Throughput Screen. 2018;21(1):41–49. doi: 10.2174/1386207320666171123201910. [DOI] [PubMed] [Google Scholar]
  • 39.Iman K., Mirza M.U., Mazhar N., Vanmeert M., Irshad I., Kamal M.A. In silico structure-based identification of novel acetylcholinesterase inhibitors against Alzheimer's disease. CNS Neurol Disord: Drug Targets. 2018;17(1):54–68. doi: 10.2174/1871527317666180115162422. [DOI] [PubMed] [Google Scholar]
  • 40.Telpoukhovskaia M.A., Patrick B.O., Rodriguez-Rodriguez C., Orvig C. In silico to in vitro screening of hydroxypyridinones as acetylcholinesterase inhibitors. Bioorg Med Chem Lett. 2016;26(6):1624–1628. doi: 10.1016/j.bmcl.2016.01.080. [DOI] [PubMed] [Google Scholar]
  • 41.Cui L., Wang Y., Liu Z., Chen H., Wang H., Zhou X., et al. Discovering new acetylcholinesterase inhibitors by mining the Buzhongyiqi decoction recipe data. J Chem Inf Model. 2015;55(11):2455–2463. doi: 10.1021/acs.jcim.5b00449. [DOI] [PubMed] [Google Scholar]
  • 42.Muthukumaran P., Rajiniraja M. MIA-QSAR based model for bioactivity prediction of flavonoid derivatives as acetylcholinesterase inhibitors. J Theor Biol. 2018;459:103–110. doi: 10.1016/j.jtbi.2018.09.030. [DOI] [PubMed] [Google Scholar]
  • 43.Jang C., Yadav D.K., Subedi L., Venkatesan R., Venkanna A., Afzal S., et al. Identification of novel acetylcholinesterase inhibitors designed by pharmacophore-based virtual screening, molecular docking and bioassay. Sci Rep. 2018;8(1) doi: 10.1038/s41598-018-33354-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.de Almeida J.R., Figueiro M., Almeida W.P., de Paula da Silva C.H.T. Discovery of novel dual acetylcholinesterase inhibitors with antifibrillogenic activity related to Alzheimer's disease. Future Med Chem. 2018;10(9):1037–1053. doi: 10.4155/fmc-2017-0201. [DOI] [PubMed] [Google Scholar]
  • 45.Jiang C.S., Ge Y.X., Cheng Z.Q., Song J.L., Wang Y.Y., Zhu K., et al. Discovery of new multifunctional selective acetylcholinesterase inhibitors: structure-based virtual screening and biological evaluation. J Comput Aided Mol Des. 2019;33(5):521–530. doi: 10.1007/s10822-019-00202-2. [DOI] [PubMed] [Google Scholar]
  • 46.Vignaux P.A., Minerali E., Lane T.R., Foil D.H., Madrid P.B., Puhl A.C., et al. The antiviral drug tilorone is a potent and selective inhibitor of acetylcholinesterase. Chem Res Toxicol. 2021;34(5):1296–1307. doi: 10.1021/acs.chemrestox.0c00466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mendez D., Gaulton A., Bento A.P., Chambers J., De Veij M., Félix E., et al. ChEMBL: towards direct deposition of bioassay data. Nucleic Acids Res. 2019;47(D1):D930–D940. doi: 10.1093/nar/gky1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Liu T., Lin Y., Wen X., Jorissen R.N., Gilson M.K. BindingDB: a web-accessible database of experimentally determined protein–ligand binding affinities. Nucleic Acids Res. 2006;35(suppl_1):D198–D201. doi: 10.1093/nar/gkl999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Vignaux P.A., Lane T.R., Urbina F., Gerlach J., Puhl A.C., Snyder S.H., et al. Validation of acetylcholinesterase inhibition machine learning models for multiple species. Chem Res Toxicol. 2023;36(2):188–201. doi: 10.1021/acs.chemrestox.2c00283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Xiong Z., Wang D., Liu X., Zhong F., Wan X., Li X., et al. Pushing the boundaries of molecular representation for drug discovery with the graph attention mechanism. J Med Chem. 2020;63(16):8749–8760. doi: 10.1021/acs.jmedchem.9b00959. [DOI] [PubMed] [Google Scholar]
  • 51.Alles G.A., Hawes R.C. Cholinesterases in the blood of man. J Biol Chem. 1940;133(2):375–390. [Google Scholar]
  • 52.Mendel B., Rudney H. On the type of cholinesterase present in brain tissue. Science. 1943;98(2539):201–202. doi: 10.1126/science.98.2539.201. [DOI] [PubMed] [Google Scholar]
  • 53.Silver A. The biology of cholinesterases. Front Biol. 1974;36:426–447. [Google Scholar]
  • 54.Darvesh S., Hopkins D.A., Geula C. Neurobiology of butyrylcholinesterase. Nat Rev Neurosci. 2003;4(2):131–138. doi: 10.1038/nrn1035. [DOI] [PubMed] [Google Scholar]
  • 55.Primo-Parmo S.L., Bartels C.F., Wiersema B., van der Spek A.F., Innis J.W., La Du B.N. Characterization of 12 silent alleles of the human butyrylcholinesterase (BCHE) gene. Am J Hum Genet. 1996;58(1):52–64. [PMC free article] [PubMed] [Google Scholar]
  • 56.Chaudhary A., Maurya P.K., Yadav B.S., Singh S., Mani A. Current therapeutic targets for Alzheimer's disease. J Biomed. 2018;3:74–84. [Google Scholar]
  • 57.Stepankova S., Komers K. Cholinesterases and cholinesterase inhibitors. Curr Enzym Inhib. 2008;4(4):160–171. [Google Scholar]
  • 58.Dubovy P., Haninec P. Non-specific cholinesterase activity of the developing peripheral nerves and its possible function in cells in intimate contact with growing axons of chick embryo. Int J Dev Neurosci. 1990;8(5):589–602. doi: 10.1016/0736-5748(90)90051-3. [DOI] [PubMed] [Google Scholar]
  • 59.Layer P.G. Cholinesterases during development of the avian nervous system. Cell Mol Neurobiol. 1991;11(1):7–33. doi: 10.1007/BF00712798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Robertson R.T., Mostamand F. Development of 'non-specific' cholinesterase-containing neurons in the dorsal thalamus of the rat. Brain Res. 1988;469(1-2):43–60. doi: 10.1016/0165-3806(88)90168-x. [DOI] [PubMed] [Google Scholar]
  • 61.Graybiel A.M., Ragsdale C.W., Jr. Pseudocholinesterase staining in the primary visual pathway of the macaque monkey. Nature. 1982;299(5882):439–442. doi: 10.1038/299439a0. [DOI] [PubMed] [Google Scholar]
  • 62.Arendt T., Bigl V., Walther F., Sonntag M. Decreased ratio of CSF acetylcholinesterase to butyrylcholinesterase activity in Alzheimer's disease. Lancet. 1984;1(8369):173. doi: 10.1016/s0140-6736(84)90116-8. [DOI] [PubMed] [Google Scholar]
  • 63.Perry E.K., Perry R.H., Blessed G., Tomlinson B.E. Changes in brain cholinesterases in senile dementia of Alzheimer type. Neuropathol Appl Neurobiol. 1978;4(4):273–277. doi: 10.1111/j.1365-2990.1978.tb00545.x. [DOI] [PubMed] [Google Scholar]
  • 64.Maelicke A., Hoeffle-Maas A., Ludwig J., Maus A., Samochocki M., Jordis U., et al. Memogain is a galantamine pro-drug having dramatically reduced adverse effects and enhanced efficacy. J Mol Neurosci. 2010;40:135–137. doi: 10.1007/s12031-009-9269-5. [DOI] [PubMed] [Google Scholar]
  • 65.Watkins P.B., Zimmerman H.J., Knapp M.J., Gracon S.I., Lewis K.W. Hepatotoxic effects of tacrine administration in patients with Alzheimer's disease. JAMA. 1994;271(13):992–998. [PubMed] [Google Scholar]
  • 66.Tayeb H.O., Yang H.D., Price B.H., Tarazi F.I. Pharmacotherapies for Alzheimer's disease: beyond cholinesterase inhibitors. Pharmacol Ther. 2012;134(1):8–25. doi: 10.1016/j.pharmthera.2011.12.002. [DOI] [PubMed] [Google Scholar]
  • 67.Li Q., Yang H., Chen Y., Sun H. Recent progress in the identification of selective butyrylcholinesterase inhibitors for Alzheimer's disease. Eur J Med Chem. 2017;132:294–309. doi: 10.1016/j.ejmech.2017.03.062. [DOI] [PubMed] [Google Scholar]
  • 68.Millard C.B., Broomfield C.A. A computer model of glycosylated human butyrylcholinesterase. Biochem Biophys Res Commun. 1992;189(3):1280–1286. doi: 10.1016/0006-291x(92)90212-4. [DOI] [PubMed] [Google Scholar]
  • 69.Nicolet Y., Lockridge O., Masson P., Fontecilla-Camps J.C., Nachon F. Crystal structure of human butyrylcholinesterase and of its complexes with substrate and products. J Biol Chem. 2003;278(42):41141–41147. doi: 10.1074/jbc.M210241200. [DOI] [PubMed] [Google Scholar]
  • 70.Saxena A., Redman A.M., Jiang X., Lockridge O., Doctor B.P. Differences in active site gorge dimensions of cholinesterases revealed by binding of inhibitors to human butyrylcholinesterase. Biochemistry. 1997;36(48):14642–14651. doi: 10.1021/bi971425+. [DOI] [PubMed] [Google Scholar]
  • 71.Ozalp M.K., Vignaux P.A., Puhl A.C., Lane T.R., Urbina F., Ekins S. Sequential contrastive and deep learning models to identify selective butyrylcholinesterase inhibitors. J Chem Inf Model. 2024;64(8):3161–3172. doi: 10.1021/acs.jcim.4c00397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Chen T., Kornblith S., Norouzi M., Hinton G., editors. International Conference on Machine Learning. PMLR; 2020. A simple framework for contrastive learning of visual representations. [Google Scholar]
  • 73.He K., Fan H., Wu Y., Xie S., Girshick R., editors. Proceedings of the IEEE/CVF Conference on Computer Vision and Pattern Recognition. 2020. Momentum contrast for unsupervised visual representation learning. [Google Scholar]
  • 74.Khosla P., Teterwak P., Wang C., Sarna A., Tian Y., Isola P., et al. Supervised contrastive learning. Adv Neural Inf Process Syst. 2020;33:18661–18673. [Google Scholar]
  • 75.Wong T.S., Li G., Li S., Gao W., Chen G., Gan S., et al. G protein-coupled receptors in neurodegenerative diseases and psychiatric disorders. Signal Transduct Targeted Ther. 2023;8(1):177. doi: 10.1038/s41392-023-01427-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Zhu S., Wu M., Huang Z., An J. Trends in application of advancing computational approaches in GPCR ligand discovery. Exp Biol Med. 2021;246(9):1011–1024. doi: 10.1177/1535370221993422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ransohoff R.M. How neuroinflammation contributes to neurodegeneration. Science. 2016;353(6301):777–783. doi: 10.1126/science.aag2590. [DOI] [PubMed] [Google Scholar]
  • 78.Koelink P.J., Overbeek S.A., Braber S., de Kruijf P., Folkerts G., Smit M.J., et al. Targeting chemokine receptors in chronic inflammatory diseases: an extensive review. Pharmacol Ther. 2012;133(1):1–18. doi: 10.1016/j.pharmthera.2011.06.008. [DOI] [PubMed] [Google Scholar]
  • 79.Cartier L., Hartley O., Dubois-Dauphin M., Krause K.H. Chemokine receptors in the central nervous system: role in brain inflammation and neurodegenerative diseases. Brain Res Brain Res Rev. 2005;48(1):16–42. doi: 10.1016/j.brainresrev.2004.07.021. [DOI] [PubMed] [Google Scholar]
  • 80.Martha S.R., Cheng Q., Fraser J.F., Gong L., Collier L.A., Davis S.M., et al. Expression of cytokines and chemokines as predictors of stroke outcomes in acute ischemic stroke. Front Neurol. 2019;10:1391. doi: 10.3389/fneur.2019.01391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Pawlik K., Mika J. Targeting members of the chemokine family as a novel approach to treating neuropathic pain. Molecules. 2023;28(15) doi: 10.3390/molecules28155766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lane T.R., Puhl A.C., Vignaux P.A., Pennypacker K.R., Ekins S. Repurposing lapatinib as a triple antagonist of chemokine receptors 3, 4, and 5. Mol Pharmacol. 2025;107(1) doi: 10.1016/j.molpha.2024.100010. [DOI] [PubMed] [Google Scholar]
  • 83.Joy M.T., Ben Assayag E., Shabashov-Stone D., Liraz-Zaltsman S., Mazzitelli J., Arenas M., et al. CCR5 is a therapeutic target for recovery after stroke and traumatic brain injury. Cell. 2019;176(5):1143–1157 e13. doi: 10.1016/j.cell.2019.01.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Jacobson K.A., Delicado E.G., Gachet C., Kennedy C., von Kugelgen I., Li B., et al. Update of P2Y receptor pharmacology: IUPHAR Review 27. Br J Pharmacol. 2020;177(11):2413–2433. doi: 10.1111/bph.15005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Koizumi S., Shigemoto-Mogami Y., Nasu-Tada K., Shinozaki Y., Ohsawa K., Tsuda M., et al. UDP acting at P2Y6 receptors is a mediator of microglial phagocytosis. Nature. 2007;446(7139):1091–1095. doi: 10.1038/nature05704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lovaszi M., Branco Haas C., Antonioli L., Pacher P., Hasko G. The role of P2Y receptors in regulating immunity and metabolism. Biochem Pharmacol. 2021;187 doi: 10.1016/j.bcp.2021.114419. [DOI] [PubMed] [Google Scholar]
  • 87.Umpierre A.D., Li B., Ayasoufi K., Simon W.L., Zhao S., Xie M., et al. Microglial P2Y(6) calcium signaling promotes phagocytosis and shapes neuroimmune responses in epileptogenesis. Neuron. 2024;112(12):1959–1977 e10. doi: 10.1016/j.neuron.2024.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Oliveira-Giacomelli A., C M.A., de Souza H.D.N., Correa-Velloso J., de Jesus Santos A.P., Baranova J., et al. P2Y6 and P2X7 receptor antagonism exerts neuroprotective/neuroregenerative effects in an animal model of Parkinson's disease. Front Cell Neurosci. 2019;13:476. doi: 10.3389/fncel.2019.00476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Milde S., van Tartwijk F.W., Vilalta A., Hornik T.C., Dundee J.M., Puigdellivol M., et al. Inflammatory neuronal loss in the substantia nigra induced by systemic lipopolysaccharide is prevented by knockout of the P2Y(6) receptor in mice. J Neuroinflammation. 2021;18(1):225. doi: 10.1186/s12974-021-02280-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Vieira R.P., Muller T., Grimm M., von Gernler V., Vetter B., Durk T., et al. Purinergic receptor type 6 contributes to airway inflammation and remodeling in experimental allergic airway inflammation. Am J Respir Crit Care Med. 2011;184(2):215–223. doi: 10.1164/rccm.201011-1762OC. [DOI] [PubMed] [Google Scholar]
  • 91.Muller T., Fay S., Vieira R.P., Karmouty-Quintana H., Cicko S., Ayata C.K., et al. P2Y(6) receptor activation promotes inflammation and tissue remodeling in pulmonary fibrosis. Front Immunol. 2017;8:1028. doi: 10.3389/fimmu.2017.01028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Jain S., Pydi S.P., Toti K.S., Robaye B., Idzko M., Gavrilova O., et al. Lack of adipocyte purinergic P2Y(6) receptor greatly improves whole body glucose homeostasis. Proc Natl Acad Sci U S A. 2020;117(48):30763–30774. doi: 10.1073/pnas.2006578117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Salem M., Lecka J., Pelletier J., Gomes Marconato D., Dumas A., Vallieres L., et al. NTPDase8 protects mice from intestinal inflammation by limiting P2Y(6) receptor activation: identification of a new pathway of inflammation for the potential treatment of IBD. Gut. 2022;71(1):43–54. doi: 10.1136/gutjnl-2020-320937. [DOI] [PubMed] [Google Scholar]
  • 94.Zhou M., Wang W., Li Y., Zhang Q., Ji H., Li H., et al. The role of P2Y(6)R in cardiovascular diseases and recent development of P2Y(6)R antagonists. Drug Discov Today. 2020;25(3):568–573. doi: 10.1016/j.drudis.2019.12.015. [DOI] [PubMed] [Google Scholar]
  • 95.Maruoka H., Barrett M.O., Ko H., Tosh D.K., Melman A., Burianek L.E., et al. Pyrimidine ribonucleotides with enhanced selectivity as P2Y(6) receptor agonists: novel 4-alkyloxyimino, (S)-methanocarba, and 5'-triphosphate gamma-ester modifications. J Med Chem. 2010;53(11):4488–4501. doi: 10.1021/jm100287t. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ginsburg-Shmuel T., Haas M., Schumann M., Reiser G., Kalid O., Stern N., et al. 5-OMe-UDP is a potent and selective P2Y(6)-receptor agonist. J Med Chem. 2010;53(4):1673–1685. doi: 10.1021/jm901450d. [DOI] [PubMed] [Google Scholar]
  • 97.Ginsburg-Shmuel T., Haas M., Grbic D., Arguin G., Nadel Y., Gendron F.P., et al. UDP made a highly promising stable, potent, and selective P2Y6-receptor agonist upon introduction of a boranophosphate moiety. Bioorg Med Chem. 2012;20(18):5483–5495. doi: 10.1016/j.bmc.2012.07.042. [DOI] [PubMed] [Google Scholar]
  • 98.Toti K.S., Jain S., Ciancetta A., Balasubramanian R., Chakraborty S., Surujdin R., et al. Pyrimidine nucleotides containing a (S)-Methanocarba ring as P2Y(6) receptor agonists. Medchemcomm. 2017;8(10):1897–1908. doi: 10.1039/c7md00397h. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Mamedova L.K., Joshi B.V., Gao Z.G., von Kugelgen I., Jacobson K.A. Diisothiocyanate derivatives as potent, insurmountable antagonists of P2Y6 nucleotide receptors. Biochem Pharmacol. 2004;67(9):1763–1770. doi: 10.1016/j.bcp.2004.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Nishiyama K., Nishimura A., Shimoda K., Tanaka T., Kato Y., Shibata T., et al. Redox-dependent internalization of the purinergic P2Y(6) receptor limits colitis progression. Sci Signal. 2022;15(716) doi: 10.1126/scisignal.abj0644. [DOI] [PubMed] [Google Scholar]
  • 101.Ito M., Egashira S.I., Yoshida K., Mineno T., Kumagai K., Kojima H., et al. Identification of novel selective P2Y(6) receptor antagonists by high-throughput screening assay. Life Sci. 2017;180:137–142. doi: 10.1016/j.lfs.2017.05.017. [DOI] [PubMed] [Google Scholar]
  • 102.Jung Y.H., Shah Q., Lewicki S.A., Pramanik A., Gopinatth V., Pelletier J., et al. Synthesis and pharmacological characterization of multiply substituted 2H-chromene derivatives as P2Y(6) receptor antagonists. Bioorg Med Chem Lett. 2022;75 doi: 10.1016/j.bmcl.2022.128981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Zhu Y., Zhou M., Cheng X., Wang H., Li Y., Guo Y., et al. Discovery of selective P2Y(6)R antagonists with high affinity and in vivo efficacy for inflammatory disease therapy. J Med Chem. 2023;66(9):6315–6332. doi: 10.1021/acs.jmedchem.3c00210. [DOI] [PubMed] [Google Scholar]
  • 104.Puhl A.C., Lewicki S.A., Gao Z.G., Pramanik A., Makarov V., Ekins S., et al. Machine learning-aided search for ligands of P2Y(6) and other P2Y receptors. Purinergic Signal. 2024;20(6):617–627. doi: 10.1007/s11302-024-10003-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Borea P.A., Gessi S., Merighi S., Vincenzi F., Varani K. Pharmacology of adenosine receptors: the state of the art. Physiol Rev. 2018;98(3):1591–1625. doi: 10.1152/physrev.00049.2017. [DOI] [PubMed] [Google Scholar]
  • 106.Borea P.A., Gessi S., Merighi S., Varani K. Adenosine as a multi-signalling guardian angel in human diseases: when, where and how does it exert its protective effects? Trends Pharmacol Sci. 2016;37(6):419–434. doi: 10.1016/j.tips.2016.02.006. [DOI] [PubMed] [Google Scholar]
  • 107.Jacobson K.A., Tosh D.K., Jain S., Gao Z.G. Historical and current adenosine receptor agonists in preclinical and clinical development. Front Cell Neurosci. 2019;13:124. doi: 10.3389/fncel.2019.00124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Effendi W.I., Nagano T., Kobayashi K., Nishimura Y. Focusing on adenosine receptors as a potential targeted therapy in human diseases. Cells. 2020;9(3) doi: 10.3390/cells9030785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Elmenhorst D., Meyer P.T., Winz O.H., Matusch A., Ermert J., Coenen H.H., et al. Sleep deprivation increases A1 adenosine receptor binding in the human brain: a positron emission tomography study. J Neurosci. 2007;27(9):2410–2415. doi: 10.1523/JNEUROSCI.5066-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Stone T.W., Ceruti S., Abbracchio M.P. Adenosine receptors and neurological disease: neuroprotection and neurodegeneration. Handb Exp Pharmacol. 2009;(193):535–587. doi: 10.1007/978-3-540-89615-9_17. [DOI] [PubMed] [Google Scholar]
  • 111.Ballesteros-Yáñez I., Castillo C.A., Merighi S., Gessi S. The role of adenosine receptors in psychostimulant addiction. Front Pharmacol. 2017;8:985. doi: 10.3389/fphar.2017.00985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Puhl A.C., Gao Z.G., Jacobson K.A., Ekins S. Machine learning for discovery of new ADORA modulators. Front Pharmacol. 2022;13 doi: 10.3389/fphar.2022.920643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Wadhwa P., Jain P., Jadhav H.R. Glycogen synthase kinase 3 (GSK3): its role and inhibitors. Curr Top Med Chem. 2020;20(17):1522–1534. doi: 10.2174/1568026620666200516153136. [DOI] [PubMed] [Google Scholar]
  • 114.Beurel E., Grieco S.F., Jope R.S. Glycogen synthase kinase-3 (GSK3): regulation, actions, and diseases. Pharmacol Ther. 2015;148:114–131. doi: 10.1016/j.pharmthera.2014.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Souder D.C., Anderson R.M. An expanding GSK3 network: implications for aging research. Geroscience. 2019;41(4):369–382. doi: 10.1007/s11357-019-00085-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Jorge-Torres O.C., Szczesna K., Roa L., Casal C., Gonzalez-Somermeyer L., Soler M., et al. Inhibition of Gsk3b reduces Nfkb1 signaling and rescues synaptic activity to improve the Rett syndrome phenotype in mecp2-knockout mice. Cell Rep. 2018;23(6):1665–1677. doi: 10.1016/j.celrep.2018.04.010. [DOI] [PubMed] [Google Scholar]
  • 117.Fuchs C., Rimondini R., Viggiano R., Trazzi S., De Franceschi M., Bartesaghi R., et al. Inhibition of GSK3beta rescues hippocampal development and learning in a mouse model of CDKL5 disorder. Neurobiol Dis. 2015;82:298–310. doi: 10.1016/j.nbd.2015.06.018. [DOI] [PubMed] [Google Scholar]
  • 118.Lee S.J., Chung Y.H., Joo K.M., Lim H.C., Jeon G.S., Kim D., et al. Age-related changes in glycogen synthase kinase 3beta (GSK3beta) immunoreactivity in the central nervous system of rats. Neurosci Lett. 2006;409(2):134–139. doi: 10.1016/j.neulet.2006.09.026. [DOI] [PubMed] [Google Scholar]
  • 119.Yuskaitis C.J., Jope R.S. Glycogen synthase kinase-3 regulates microglial migration, inflammation, and inflammation-induced neurotoxicity. Cell Signal. 2009;21(2):264–273. doi: 10.1016/j.cellsig.2008.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Bernaus A., Blanco S., Sevilla A. Glia crosstalk in neuroinflammatory diseases. Front Cell Neurosci. 2020;14:209. doi: 10.3389/fncel.2020.00209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Gianferrara T., Cescon E., Grieco I., Spalluto G., Federico S. Glycogen synthase kinase 3β involvement in neuroinflammation and neurodegenerative diseases. Curr Med Chem. 2022;29(27):4631–4697. doi: 10.2174/0929867329666220216113517. [DOI] [PubMed] [Google Scholar]
  • 122.Valvezan A.J., Klein P.S. GSK-3 and Wnt signaling in neurogenesis and bipolar disorder. Front Mol Neurosci. 2012;5:1. doi: 10.3389/fnmol.2012.00001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Arciniegas Ruiz S.M., Eldar-Finkelman H. Glycogen synthase kinase-3 inhibitors: preclinical and clinical focus on CNS-A decade onward. Front Mol Neurosci. 2021;14 doi: 10.3389/fnmol.2021.792364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Leroy K., Yilmaz Z., Brion J.P. Increased level of active GSK-3beta in Alzheimer's disease and accumulation in argyrophilic grains and in neurones at different stages of neurofibrillary degeneration. Neuropathol Appl Neurobiol. 2007;33(1):43–55. doi: 10.1111/j.1365-2990.2006.00795.x. [DOI] [PubMed] [Google Scholar]
  • 125.Pei J.J., Tanaka T., Tung Y.C., Braak E., Iqbal K., Grundke-Iqbal I. Distribution, levels, and activity of glycogen synthase kinase-3 in the Alzheimer disease brain. J Neuropathol Exp Neurol. 1997;56(1):70–78. doi: 10.1097/00005072-199701000-00007. [DOI] [PubMed] [Google Scholar]
  • 126.Ishiguro K., Omori A., Takamatsu M., Sato K., Arioka M., Uchida T., et al. Phosphorylation sites on tau by tau protein kinase I, a bovine derived kinase generating an epitope of paired helical filaments. Neurosci Lett. 1992;148(1-2):202–206. doi: 10.1016/0304-3940(92)90839-y. [DOI] [PubMed] [Google Scholar]
  • 127.Hanger D.P., Hughes K., Woodgett J.R., Brion J.P., Anderton B.H. Glycogen synthase kinase-3 induces Alzheimer's disease-like phosphorylation of tau: generation of paired helical filament epitopes and neuronal localisation of the kinase. Neurosci Lett. 1992;147(1):58–62. doi: 10.1016/0304-3940(92)90774-2. [DOI] [PubMed] [Google Scholar]
  • 128.Lovestone S., Reynolds C.H., Latimer D., Davis D.R., Anderton B.H., Gallo J.M., et al. Alzheimer's disease-like phosphorylation of the microtubule-associated protein tau by glycogen synthase kinase-3 in transfected mammalian cells. Curr Biol. 1994;4(12):1077–1086. doi: 10.1016/s0960-9822(00)00246-3. [DOI] [PubMed] [Google Scholar]
  • 129.Vanden Dries V., Stygelbout V., Pierrot N., Yilmaz Z., Suain V., De Decker R., et al. Amyloid precursor protein reduction enhances the formation of neurofibrillary tangles in a mutant tau transgenic mouse model. Neurobiol Aging. 2017;55:202–212. doi: 10.1016/j.neurobiolaging.2017.03.031. [DOI] [PubMed] [Google Scholar]
  • 130.Engel T., Goni-Oliver P., Lucas J.J., Avila J., Hernandez F. Chronic lithium administration to FTDP-17 tau and GSK-3beta overexpressing mice prevents tau hyperphosphorylation and neurofibrillary tangle formation, but pre-formed neurofibrillary tangles do not revert. J Neurochem. 2006;99(6):1445–1455. doi: 10.1111/j.1471-4159.2006.04139.x. [DOI] [PubMed] [Google Scholar]
  • 131.Uemura K., Kuzuya A., Shimozono Y., Aoyagi N., Ando K., Shimohama S., et al. GSK3beta activity modifies the localization and function of presenilin 1. J Biol Chem. 2007;282(21):15823–15832. doi: 10.1074/jbc.M610708200. [DOI] [PubMed] [Google Scholar]
  • 132.Ly P.T., Wu Y., Zou H., Wang R., Zhou W., Kinoshita A., et al. Inhibition of GSK3beta-mediated BACE1 expression reduces Alzheimer-associated phenotypes. J Clin Investig. 2013;123(1):224–235. doi: 10.1172/JCI64516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Maqbool M., Mobashir M., Hoda N. Pivotal role of glycogen synthase kinase-3: a therapeutic target for Alzheimer's disease. Eur J Med Chem. 2016;107:63–81. doi: 10.1016/j.ejmech.2015.10.018. [DOI] [PubMed] [Google Scholar]
  • 134.Duda P., Wisniewski J., Wojtowicz T., Wojcicka O., Jaskiewicz M., Drulis-Fajdasz D., et al. Targeting GSK3 signaling as a potential therapy of neurodegenerative diseases and aging. Expert Opin Ther Targets. 2018;22(10):833–848. doi: 10.1080/14728222.2018.1526925. [DOI] [PubMed] [Google Scholar]
  • 135.Walz A., Ugolkov A., Chandra S., Kozikowski A., Carneiro B.A., O'Halloran T.V., et al. Molecular pathways: revisiting glycogen synthase kinase-3beta as a target for the treatment of cancer. Clin Cancer Res. 2017;23(8):1891–1897. doi: 10.1158/1078-0432.CCR-15-2240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Ryder J., Su Y., Liu F., Li B., Zhou Y., Ni B. Divergent roles of GSK3 and CDK5 in APP processing. Biochem Biophys Res Commun. 2003;312(4):922–929. doi: 10.1016/j.bbrc.2003.11.014. [DOI] [PubMed] [Google Scholar]
  • 137.Sereno L., Coma M., Rodriguez M., Sanchez-Ferrer P., Sanchez M.B., Gich I., et al. A novel GSK-3beta inhibitor reduces Alzheimer's pathology and rescues neuronal loss in vivo. Neurobiol Dis. 2009;35(3):359–367. doi: 10.1016/j.nbd.2009.05.025. [DOI] [PubMed] [Google Scholar]
  • 138.Fukunaga K., Sakai D., Watanabe K., Nakayama K., Kohara T., Tanaka H., et al. Discovery of novel 2-(alkylmorpholin-4-yl)-6-(3-fluoropyridin-4-yl)-pyrimidin-4(3H)-ones as orally-active GSK-3beta inhibitors for Alzheimer's disease. Bioorg Med Chem Lett. 2015;25(5):1086–1091. doi: 10.1016/j.bmcl.2015.01.005. [DOI] [PubMed] [Google Scholar]
  • 139.Luo G., Chen L., Burton C.R., Xiao H., Sivaprakasam P., Krause C.M., et al. Discovery of isonicotinamides as highly selective, brain penetrable, and orally active glycogen synthase kinase-3 inhibitors. J Med Chem. 2016;59(3):1041–1051. doi: 10.1021/acs.jmedchem.5b01550. [DOI] [PubMed] [Google Scholar]
  • 140.Berg S., Bergh M., Hellberg S., Hogdin K., Lo-Alfredsson Y., Soderman P., et al. Discovery of novel potent and highly selective glycogen synthase kinase-3beta (GSK3beta) inhibitors for Alzheimer's disease: design, synthesis, and characterization of pyrazines. J Med Chem. 2012;55(21):9107–9119. doi: 10.1021/jm201724m. [DOI] [PubMed] [Google Scholar]
  • 141.Varela-Nallar L., Arredondo S.B., Tapia-Rojas C., Hancke J., Inestrosa N.C. Andrographolide stimulates neurogenesis in the adult Hippocampus. Neural Plast. 2015;2015 doi: 10.1155/2015/935403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Griebel G., Stemmelin J., Lopez-Grancha M., Boulay D., Boquet G., Slowinski F., et al. The selective GSK3 inhibitor, SAR502250, displays neuroprotective activity and attenuates behavioral impairments in models of neuropsychiatric symptoms of Alzheimer's disease in rodents. Sci Rep. 2019;9(1) doi: 10.1038/s41598-019-54557-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Huang H.J., Chen S.L., Huang H.Y., Sun Y.C., Lee G.C., Lee-Chen G.J., et al. Chronic low dose of AM404 ameliorates the cognitive impairment and pathological features in hyperglycemic 3xTg-AD mice. Psychopharmacology (Berl). 2019;236(2):763–773. doi: 10.1007/s00213-018-5108-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Liang S.H., Chen J.M., Normandin M.D., Chang J.S., Chang G.C., Taylor C.K., et al. Discovery of a highly selective glycogen synthase kinase-3 inhibitor (PF-04802367) that modulates tau phosphorylation in the brain: translation for PET neuroimaging. Angew Chem Int Ed Engl. 2016;55(33):9601–9605. doi: 10.1002/anie.201603797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Matsunaga S., Fujishiro H., Takechi H. Efficacy and safety of glycogen synthase kinase 3 inhibitors for alzheimer's disease: a systematic review and meta-analysis. J Alzheimers Dis. 2019;69(4):1031–1039. doi: 10.3233/JAD-190256. [DOI] [PubMed] [Google Scholar]
  • 146.Kuroki H., Anraku T., Kazama A., Bilim V., Tasaki M., Schmitt D., et al. 9-ING-41, a small molecule inhibitor of GSK-3beta, potentiates the effects of anticancer therapeutics in bladder cancer. Sci Rep. 2019;9(1) doi: 10.1038/s41598-019-56461-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Vignaux P.A., Minerali E., Foil D.H., Puhl A.C., Ekins S. Machine learning for discovery of GSK3beta inhibitors. ACS Omega. 2020;5(41):26551–26561. doi: 10.1021/acsomega.0c03302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Jones T., Raman R., Puhl A.C., Lane T.R., Riabova O., Kazakova E., et al. Discovery of dual targeting GSK-3beta/HIV-1 reverse transcriptase inhibitors as neuroprotective antiviral agents. ACS Chem Neurosci. 2025;16(1):77–84. doi: 10.1021/acschemneuro.4c00725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Bennett D.L., Clark A.J., Huang J., Waxman S.G., Dib-Hajj S.D. The role of voltage-gated sodium channels in pain signaling. Physiol Rev. 2019;99(2):1079–1151. doi: 10.1152/physrev.00052.2017. [DOI] [PubMed] [Google Scholar]
  • 150.Kaluza L., Meents J.E., Hampl M., Rosseler C., Hautvast P.A.I., Detro-Dassen S., et al. Loss-of-function of Nav1.8/D1639N linked to human pain can be rescued by lidocaine. Pflügers Archiv. 2018;470(12):1787–1801. doi: 10.1007/s00424-018-2189-x. [DOI] [PubMed] [Google Scholar]
  • 151.Gonzalez-Lopez E., Imamura Kawasawa Y., Walter V., Zhang L., Koltun W.A., Huang X., et al. Homozygosity for the SCN10A polymorphism rs6795970 is associated with hypoalgesic inflammatory bowel disease phenotype. Front Med. 2018;5:324. doi: 10.3389/fmed.2018.00324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Entrez Gene Sodium channel [SCN10A sodium voltage-gated channel alpha subunit. https://www.ncbi.nlm.nih.gov/gene?Db=gene&Cmd=ShowDetailView&TermToSearch=6336
  • 153.Catterall W.A., Perez-Reyes E., Snutch T.P., Striessnig J. International Union of Pharmacology. XLVIII. Nomenclature and structure-function relationships of voltage-gated calcium channels. Pharmacol Rev. 2005;57(4):411–425. doi: 10.1124/pr.57.4.5. [DOI] [PubMed] [Google Scholar]
  • 154.Plummer N.W., Meisler M.H. Evolution and diversity of mammalian sodium channel genes. Genomics. 1999;57(2):323–331. doi: 10.1006/geno.1998.5735. [DOI] [PubMed] [Google Scholar]
  • 155.Rabert D.K., Koch B.D., Ilnicka M., Obernolte R.A., Naylor S.L., Herman R.C., et al. A tetrodotoxin-resistant voltage-gated sodium channel from human dorsal root ganglia, hPN3/SCN10A. Pain. 1998;78(2):107–114. doi: 10.1016/S0304-3959(98)00120-1. [DOI] [PubMed] [Google Scholar]
  • 156.Akopian A.N., Sivilotti L., Wood J.N. A tetrodotoxin-resistant voltage-gated sodium channel expressed by sensory neurons. Nature. 1996;379(6562):257–262. doi: 10.1038/379257a0. [DOI] [PubMed] [Google Scholar]
  • 157.Akopian A.N.S.V., England S., Okuse K., Ogata N., Ure J., Smith A., et al. The tetrodotoxin-resistant sodium channel SNS has a specialized function in pain pathways. Nat Neurosci. 1999;2:541–548. doi: 10.1038/9195. [DOI] [PubMed] [Google Scholar]
  • 158.Cummins T.R., Sheets P.L., Waxman S.G. The roles of sodium channels in nociception: implications for mechanisms of pain. Pain. 2007;131(3):243–257. doi: 10.1016/j.pain.2007.07.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Nardi A., Damann N., Hertrampf T., Kless A. Advances in targeting voltage-gated sodium channels with small molecules. ChemMedChem. 2012;7(10):1712–1740. doi: 10.1002/cmdc.201200298. [DOI] [PubMed] [Google Scholar]
  • 160.Anon. PF-06305591 [Available from: https://clinicaltrials.gov/ct2/results?cond=&term=PF06305591&cntry1=&state1=&SearchAll=Search+all+studies&recrs=.
  • 161.Osteen J.D., Immani S., Tapley T.L., Indersmitten T., Hurst N.W., Healey T., et al. Pharmacology and mechanism of action of suzetrigine, a potent and selective Na(V)1.8 pain signal inhibitor for the treatment of moderate to severe pain. Pain Ther. 2025;14(2):655–674. doi: 10.1007/s40122-024-00697-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Jones J., Correll D.J., Lechner S.M., Jazic I., Miao X., Shaw D., et al. Selective inhibition of Na(V)1.8 with VX-548 for acute pain. N Engl J Med. 2023;389(5):393–405. doi: 10.1056/NEJMoa2209870. [DOI] [PubMed] [Google Scholar]
  • 163.Rannals M.D., Hamersky G.R., Page S.C., Campbell M.N., Briley A., Gallo R.A., et al. Psychiatric risk gene transcription factor 4 regulates intrinsic excitability of prefrontal neurons via repression of SCN10a and KCNQ1. Neuron. 2016;90(1):43–55. doi: 10.1016/j.neuron.2016.02.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Ekins S., Gerlach J., Zorn K.M., Antonio B.M., Lin Z., Gerlach A. Repurposing approved drugs as inhibitors of K(v)7.1 and Na(v)1.8 to treat Pitt Hopkins syndrome. Pharm Res. 2019;36(9):137. doi: 10.1007/s11095-019-2671-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Fogel D.B. Factors associated with clinical trials that fail and opportunities for improving the likelihood of success: a review. Cont Clin Trial Comm. 2018;11:156–164. doi: 10.1016/j.conctc.2018.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Bhhatarai B., Walters W.P., Hop C., Lanza G., Ekins S. Opportunities and challenges using artificial intelligence in ADME/Tox. Nat Mater. 2019;18(5):418–422. doi: 10.1038/s41563-019-0332-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Salinsky L.M., Merritt C.R., Zamora J.C., Giacomini J.L., Anastasio N.C., Cunningham K.A. μ-opioid receptor agonists and psychedelics: pharmacological opportunities and challenges. Front Pharmacol. 2023;14 doi: 10.3389/fphar.2023.1239159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Jones G., Ricard J.A., Lipson J., Nock M.K. Associations between classic psychedelics and opioid use disorder in a nationally-representative U.S. adult sample. Sci Rep. 2022;12(1):4099. doi: 10.1038/s41598-022-08085-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Peters J., Olson D.E. Engineering safer psychedelics for treating addiction. Neuroscience Insights. 2021;16 doi: 10.1177/26331055211033847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Dos Santos R.G., Hallak J.E.C. Ayahuasca, an ancient substance with traditional and contemporary use in neuropsychiatry and neuroscience. Epilepsy Behav. 2019 doi: 10.1016/j.yebeh.2019.04.053. [DOI] [PubMed] [Google Scholar]
  • 171.Dong C., Ly C., Dunlap L.E., Vargas M.V., Sun J., Hwang I.W., et al. Psychedelic-inspired drug discovery using an engineered biosensor. Cell. 2021;184(10):2779–2792 e18. doi: 10.1016/j.cell.2021.03.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Shulgin A., Shulgin A. Transform press; Berkeley, CA: 1990. PIHKAL: a chemical love story. [Google Scholar]
  • 173.Shulgin A., Shulgin A. Transform Press; Berkeley, CA: 2002. TIHKAL: the continuation. [Google Scholar]
  • 174.Urbina F., Jones T., Harris J.S., Snyder S.H., Lane T.R., Ekins S. Predicting the hallucinogenic potential of molecules using artificial intelligence. ACS Chem Neurosci. 2024;15(16):3078–3089. doi: 10.1021/acschemneuro.4c00405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Duan W., Cao D., Wang S., Cheng J. Serotonin 2A receptor (5-HT(2A)R) agonists: psychedelics and non-hallucinogenic analogues as emerging antidepressants. Chem Rev. 2024;124(1):124–163. doi: 10.1021/acs.chemrev.3c00375. [DOI] [PubMed] [Google Scholar]
  • 176.Lane T.R., Snyder S.H., Harris J.S., Urbina F., Ekins S. 2025. Adverse outcome pathway and machine learning to predict drug induced seizure liability. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Zhang H., Li W., Xie Y., Wang W.J., Li L.L., Yang S.Y. Rapid and accurate assessment of seizure liability of drugs by using an optimal support vector machine method. Toxicol Vitro. 2011;25(8):1848–1854. doi: 10.1016/j.tiv.2011.05.015. [DOI] [PubMed] [Google Scholar]
  • 178.Wang Q., Cohen J.D., Yukawa T., Estrella H., Leonard C., Nunes J., et al. Assessment of a 3D neural spheroid model to detect pharmaceutical-induced neurotoxicity. ALTEX. 2022;39(4):560–582. doi: 10.14573/altex.2112221. [DOI] [PubMed] [Google Scholar]
  • 179.Matsuda N., Odawara A., Kinoshita K., Okamura A., Shirakawa T., Suzuki I. Raster plots machine learning to predict the seizure liability of drugs and to identify drugs. Sci Rep. 2022;12(1):2281. doi: 10.1038/s41598-022-05697-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Kreir M., Van Deuren B., Versweyveld S., De Bondt A., Van den Wyngaert I., Van der Linde H., et al. Do in vitro assays in rat primary neurons predict drug-induced seizure liability in humans? Toxicol Appl Pharmacol. 2018;346:45–57. doi: 10.1016/j.taap.2018.03.028. [DOI] [PubMed] [Google Scholar]
  • 181.Bradley J.A., Luithardt H.H., Metea M.R., Strock C.J. In vitro screening for seizure liability using microelectrode array technology. Toxicol Sci. 2018;163(1):240–253. doi: 10.1093/toxsci/kfy029. [DOI] [PubMed] [Google Scholar]
  • 182.Gao M., Igata H., Takeuchi A., Sato K., Ikegaya Y. Machine learning-based prediction of adverse drug effects: an example of seizure-inducing compounds. J Pharmacol Sci. 2017;133(2):70–78. doi: 10.1016/j.jphs.2017.01.003. [DOI] [PubMed] [Google Scholar]
  • 183.Koseki N., Deguchi J., Yamashita A., Miyawaki I., Funabashi H. Establishment of a novel experimental protocol for drug-induced seizure liability screening based on a locomotor activity assay in zebrafish. J Toxicol Sci. 2014;39(4):579–600. doi: 10.2131/jts.39.579. [DOI] [PubMed] [Google Scholar]
  • 184.Winter M.J., Redfern W.S., Hayfield A.J., Owen S.F., Valentin J.P., Hutchinson T.H. Validation of a larval zebrafish locomotor assay for assessing the seizure liability of early-stage development drugs. J Pharmacol Toxicol Methods. 2008;57(3):176–187. doi: 10.1016/j.vascn.2008.01.004. [DOI] [PubMed] [Google Scholar]
  • 185.Hogberg-Durdock H.H.J., Borrel A., Karmaus A., Lane T., Unnikrishnan A., Urbina F., et al., editors. "Identifying seizure-inducing biological targets: a collaborative approach using AOP networks, In Vitro assays, and computational tools", the toxicologist, a supplement to toxicological sciences, Abstract #1045. 2024. [Google Scholar]
  • 186.Chen Y., Liu L. Modern methods for delivery of drugs across the blood-brain barrier. Adv Drug Deliv Rev. 2012;64(7):640–665. doi: 10.1016/j.addr.2011.11.010. [DOI] [PubMed] [Google Scholar]
  • 187.Ekins S., Kortagere S., Iyer M., Reschly E.J., Lill M.A., Redinbo M.R., et al. Challenges predicting ligand-receptor interactions of promiscuous proteins: the nuclear receptor PXR. PLoS Comput Biol. 2009;5(12) doi: 10.1371/journal.pcbi.1000594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Rankovic Z. CNS physicochemical property space shaped by a diverse set of molecules with experimentally determined exposure in the mouse brain. J Med Chem. 2017;60(14):5943–5954. doi: 10.1021/acs.jmedchem.6b01469. [DOI] [PubMed] [Google Scholar]
  • 189.Rankovic Z. Retraction of "CNS physicochemical property space shaped by a diverse set of molecules with experimentally determined exposure in the mouse brain". J Med Chem. 2019;62(3):1699. doi: 10.1021/acs.jmedchem.8b01388. [DOI] [PubMed] [Google Scholar]
  • 190.Zhang X., Liu T., Fan X., Ai N. In silico modeling on ADME properties of natural products: classification models for blood-brain barrier permeability, its application to traditional Chinese medicine and in vitro experimental validation. J Mol Graph Model. 2017;75:347–354. doi: 10.1016/j.jmgm.2017.05.021. [DOI] [PubMed] [Google Scholar]
  • 191.Zhang D., Xiao J., Zhou N., Zheng M., Luo X., Jiang H., et al. A genetic algorithm based support vector machine model for blood-brain barrier penetration prediction. BioMed Res Int. 2015;2015 doi: 10.1155/2015/292683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Yuan Y., Zheng F., Zhan C.G. Improved prediction of blood-brain barrier permeability through machine learning with combined use of molecular property-based descriptors and fingerprints. AAPS J. 2018;20(3):54. doi: 10.1208/s12248-018-0215-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Plisson F., Piggott A.M. Predicting Blood(-)Brain barrier permeability of marine-derived kinase inhibitors using ensemble classifiers reveals potential hits for neurodegenerative disorders. Mar Drugs. 2019;17(2) doi: 10.3390/md17020081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Miao R., Xia L.Y., Chen H.H., Huang H.H., Liang Y. Improved classification of blood-brain-barrier drugs using deep learning. Sci Rep. 2019;9(1):8802. doi: 10.1038/s41598-019-44773-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Alsenan S., Al-Turaiki I., Hafez A. A Recurrent Neural Network model to predict blood-brain barrier permeability. Comput Biol Chem. 2020;89 doi: 10.1016/j.compbiolchem.2020.107377. [DOI] [PubMed] [Google Scholar]
  • 196.Shaker B., Yu M.S., Song J.S., Ahn S., Ryu J.Y., Oh K.S., et al. LightBBB: computational prediction model of blood-brain-barrier penetration based on LightGBM. Bioinformatics. 2021;37(8):1135–1139. doi: 10.1093/bioinformatics/btaa918. [DOI] [PubMed] [Google Scholar]
  • 197.Majumdar S., Basak S.C., Lungu C.N., Diudea M.V., Grunwald G.D. Finding needles in a Haystack: determining key molecular descriptors associated with the blood-brain barrier entry of chemical compounds using machine learning. Mol Inform. 2019;38(8-9) doi: 10.1002/minf.201800164. [DOI] [PubMed] [Google Scholar]
  • 198.Wager T.T., Hou X., Verhoest P.R., Villalobos A. Moving beyond rules: the development of a central nervous system multiparameter optimization (CNS MPO) approach to enable alignment of druglike properties. ACS Chem Neurosci. 2010;1(6):435–449. doi: 10.1021/cn100008c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Wager T.T., Hou X., Verhoest P.R., Villalobos A. Central nervous system multiparameter optimization desirability: application in drug discovery. ACS Chem Neurosci. 2016;7(6):767–775. doi: 10.1021/acschemneuro.6b00029. [DOI] [PubMed] [Google Scholar]
  • 200.Urbina F., Zorn K.M., Brunner D., Ekins S. Comparing the Pfizer central nervous system multiparameter optimization calculator and a BBB machine learning model. ACS Chem Neurosci. 2021;12(12):2247–2253. doi: 10.1021/acschemneuro.1c00265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Vatansever S., Schlessinger A., Wacker D., Kaniskan H.U., Jin J., Zhou M.M., et al. Artificial intelligence and machine learning-aided drug discovery in central nervous system diseases: state-of-the-arts and future directions. Med Res Rev. 2021;41(3):1427–1473. doi: 10.1002/med.21764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Himmelstein D.S., Lizee A., Hessler C., Brueggeman L., Chen S.L., Hadley D., et al. Systematic integration of biomedical knowledge prioritizes drugs for repurposing. eLife. 2017;6 doi: 10.7554/eLife.26726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Luscher Dias T., Schuch V., Beltrao-Braga P.C.B., Martins-de-Souza D., Brentani H.P., Franco G.R., et al. Drug repositioning for psychiatric and neurological disorders through a network medicine approach. Transl Psychiatry. 2020;10(1):141. doi: 10.1038/s41398-020-0827-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Xu J., Mao C., Hou Y., Luo Y., Binder J.L., Zhou Y., et al. Interpretable deep learning translation of GWAS and multi-omics findings to identify pathobiology and drug repurposing in Alzheimer's disease. Cell Rep. 2022;41(9) doi: 10.1016/j.celrep.2022.111717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Tandra G., Yoone A., Mathew R., Wang M., Hales C.M., Mitchell C.S. Literature-based discovery predicts antihistamines are a promising repurposed adjuvant therapy for Parkinson's disease. Int J Mol Sci. 2023;24(15) doi: 10.3390/ijms241512339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Vicidomini C., Fontanella F., D'Alessandro T., Roviello G.N. A survey on computational methods in drug discovery for neurodegenerative diseases. Biomolecules. 2024;14(10) doi: 10.3390/biom14101330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Anstine D.M., Isayev O. Generative models as an emerging paradigm in the chemical sciences. J Am Chem Soc. 2023;145(16):8736–8750. doi: 10.1021/jacs.2c13467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Olivecrona M., Blaschke T., Engkvist O., Chen H. Molecular de-novo design through deep reinforcement learning. J Cheminf. 2017;9(1):48. doi: 10.1186/s13321-017-0235-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Segler M.H.S., Kogej T., Tyrchan C., Waller M.P. Generating focused molecule libraries for drug discovery with recurrent neural networks. ACS Cent Sci. 2018;4(1):120–131. doi: 10.1021/acscentsci.7b00512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Prykhodko O., Johansson S.V., Kotsias P.C., Arus-Pous J., Bjerrum E.J., Engkvist O., et al. A de novo molecular generation method using latent vector based generative adversarial network. J Cheminf. 2019;11(1):74. doi: 10.1186/s13321-019-0397-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Gomez-Bombarelli R., Wei J.N., Duvenaud D., Hernandez-Lobato J.M., Sanchez-Lengeling B., Sheberla D., et al. Automatic chemical design using a data-driven continuous representation of molecules. ACS Cent Sci. 2018;4(2):268–276. doi: 10.1021/acscentsci.7b00572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Kang S.G., Morrone J.A., Weber J.K., Cornell W.D. Analysis of training and seed bias in small molecules generated with a conditional graph-based variational autoencoder horizontal line insights for practical AI-driven molecule generation. J Chem Inf Model. 2022;62(4):801–816. doi: 10.1021/acs.jcim.1c01545. [DOI] [PubMed] [Google Scholar]
  • 213.Hochreiter S., Schmidhuber J. Long short-term memory. Neural Comput. 1997;9:1735–1780. doi: 10.1162/neco.1997.9.8.1735. [DOI] [PubMed] [Google Scholar]
  • 214.Blaschke T., Olivecrona M., Engkvist O., Bajorath J., Chen H. Application of generative autoencoder in de novo molecular design. Mol Inform. 2018;37(1-2) doi: 10.1002/minf.201700123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Sanchez-Lengeling B., Outeiral C., Guimaraes G.L., Aspuru-Guzik A. Optimizing distributions over molecular space. An objective-reinforced generative adversarial network for inverse-design chemistry (ORGANIC) 2017. https://chemrxiv.org/engage/chemrxiv/article-details/60c73d91702a9beea7189bc2
  • 216.Winter R., Montanari F., Steffen A., Briem H., Noé F., Clevert D.-A. Efficient multi-objective molecular optimization in a continuous latent space. Chem Sci. 2019;10(34):8016–8024. doi: 10.1039/c9sc01928f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Krenn M., Häse F., Nigam A., Friederich P., Aspuru-Guzik A. Self-referencing embedded strings (SELFIES): a 100% robust molecular string representation. Mach Learn: Sci Technol. 2020;1(4) [Google Scholar]
  • 218.Jin W, Barzilay R, Jaakola T. Junction tree variational autoencoder for molecular graph generation 2019 [Available from: https://arxiv.org/pdf/1802.04364.pdf.
  • 219.Maziarka L., Pocha A., Kaczmarczyk J., Rataj K., Danel T., Warchol M. Mol-CycleGAN: a generative model for molecular optimization. J Cheminf. 2020;12:2. doi: 10.1186/s13321-019-0404-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Gupta A., Muller A.T., Huisman B.J.H., Fuchs J.A., Schneider P., Schneider G. Erratum: generative recurrent networks for de novo drug design. Mol Inform. 2018;37(1-2) doi: 10.1002/minf.201700111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Bjerrum EJ, Threlfall R. Molecular generation with recurrent neural networks (RNNs). arXiv. 2017;1705.04612.
  • 222.Zhavoronkov A., Ivanenkov Y.A., Aliper A., Veselov M.S., Aladinskiy V.A., Aladinskaya A.V., et al. Deep learning enables rapid identification of potent DDR1 kinase inhibitors. Nat Biotechnol. 2019;37(9):1038–1040. doi: 10.1038/s41587-019-0224-x. [DOI] [PubMed] [Google Scholar]
  • 223.Putin E., Asadulaev A., Vanhaelen Q., Ivanenkov Y., Aladinskaya A.V., Aliper A., et al. Adversarial Threshold Neural Computer for Molecular de Novo Design. Mol Pharm. 2018;15(10):4386–4397. doi: 10.1021/acs.molpharmaceut.7b01137. [DOI] [PubMed] [Google Scholar]
  • 224.Ai C., Yang H., Liu X., Dong R., Ding Y., Guo F. MTMol-GPT: de novo multi-target molecular generation with transformer-based generative adversarial imitation learning. PLoS Comput Biol. 2024;20(6) doi: 10.1371/journal.pcbi.1012229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Singh V., Singh S.K., Sharma R. A novel framework based on explainable AI and genetic algorithms for designing neurological medicines. Sci Rep. 2024;14(1) doi: 10.1038/s41598-024-63561-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Horne R.I., Andrzejewska E.A., Alam P., Brotzakis Z.F., Srivastava A., Aubert A., et al. Discovery of potent inhibitors of alpha-synuclein aggregation using structure-based iterative learning. Nat Chem Biol. 2024;20(5):634–645. doi: 10.1038/s41589-024-01580-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Macalino S.J.Y., Basith S., Clavio N.A.B., Chang H., Kang S., Choi S. Evolution of in silico strategies for protein-protein interaction drug discovery. Molecules. 2018;23(8) doi: 10.3390/molecules23081963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Mulpuri R.P., Konda N., Gadde S.T., Amalakanti S., Valiveti S.C. Artificial intelligence and machine learning in neuroregeneration: a systematic review. Cureus. 2024;16(5) doi: 10.7759/cureus.61400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Rudroff T. Artificial intelligence as a replacement for animal experiments in neurology: potential, progress, and challenges. Neurol Int. 2024;16(4):805–820. doi: 10.3390/neurolint16040060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Ireland D., Rabeler C., Rao S., Richardson R.J., Collins E.S. Distinguishing classes of neuroactive drugs based on computational physicochemical properties and experimental phenotypic profiling in planarians. PLoS One. 2025;20(1) doi: 10.1371/journal.pone.0315394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Geldenhuys W.J., Bloomquist J.R. Development of an a priori computational approach for brain uptake of compounds in an insect model system. Bioorg Med Chem Lett. 2021;40 doi: 10.1016/j.bmcl.2021.127930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.de Jong J., Cutcutache I., Page M., Elmoufti S., Dilley C., Frohlich H., et al. Towards realizing the vision of precision medicine: AI based prediction of clinical drug response. Brain. 2021;144(6):1738–1750. doi: 10.1093/brain/awab108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Underwood B.R., Lourida I., Gong J., Tamburin S., Tang E.Y.H., Sidhom E., et al. Data-driven discovery of associations between prescribed drugs and dementia risk: a systematic review. Alzheimer's Dement. 2025;11(1) doi: 10.1002/trc2.70037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Doherty T., Yao Z., Khleifat A.A.L., Tantiangco H., Tamburin S., Albertyn C., et al. Artificial intelligence for dementia drug discovery and trials optimization. Alzheimer's Dement. 2023;19(12):5922–5933. doi: 10.1002/alz.13428. [DOI] [PubMed] [Google Scholar]
  • 235.Akras Z., Jing J., Westover M.B., Zafar S.F. Using artificial intelligence to optimize anti-seizure treatment and EEG-guided decisions in severe brain injury. Neurotherapeutics. 2025;22(1) doi: 10.1016/j.neurot.2025.e00524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Rodriguez S., Hug C., Todorov P., Moret N., Boswell S.A., Evans K., et al. Machine learning identifies candidates for drug repurposing in Alzheimer's disease. Nat Commun. 2021;12(1):1033. doi: 10.1038/s41467-021-21330-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Roberds S.L., Filippov I., Alexandrov V., Hanania T., Brunner D. Rapid, computer vision-enabled murine screening system identifies neuropharmacological potential of two new mechanisms. Front Neurosci. 2011;5:103. doi: 10.3389/fnins.2011.00103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Akere M.T., Zajac K.K., Bretz J.D., Madhavaram A.R., Horton A.C., Schiefer I.T. Real-time analysis of neuronal cell cultures for CNS drug discovery. Brain Sci. 2024;14(8) doi: 10.3390/brainsci14080770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Luo X., Ding Y., Cao Y., Liu Z., Zhang W., Zeng S., et al. Few-shot meta-learning applied to whole brain activity maps improves systems neuropharmacology and drug discovery. iScience. 2024;27(10) doi: 10.1016/j.isci.2024.110875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Lee S., Weiss T., Buhler M., Mena J., Lottenbach Z., Wegmann R., et al. High-throughput identification of repurposable neuroactive drugs with potent anti-glioblastoma activity. Nat Med. 2024;30(11):3196–3208. doi: 10.1038/s41591-024-03224-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Tan R.K., Liu Y., Xie L. Reinforcement learning for systems pharmacology-oriented and personalized drug design. Expet Opin Drug Discov. 2022;17(8):849–863. doi: 10.1080/17460441.2022.2072288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Liu D., Liu D., Sheng K., Cheng Z., Liu Z., Qiao Y., et al. STNGS: a deep scaffold learning-driven generation and screening framework for discovering potential novel psychoactive substances. Briefings Bioinf. 2024;26(1) doi: 10.1093/bib/bbae690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Urbina F., Lentzos F., Invernizzi C., Ekins S. Dual use of artificial intelligence-powered drug discovery. Nat Mach Intell. 2022;4(3):189–191. doi: 10.1038/s42256-022-00465-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Kosten T.R., Baxter L.E. Review article: effective management of opioid withdrawal symptoms: a gateway to opioid dependence treatment. Am J Addict. 2019;28(2):55–62. doi: 10.1111/ajad.12862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Wesson D.R., Ling W. The clinical opiate withdrawal scale (COWS) J Psychoact Drugs. 2003;35(2):253–259. doi: 10.1080/02791072.2003.10400007. [DOI] [PubMed] [Google Scholar]
  • 246.Cicero T.J., Ellis M.S. The prescription opioid epidemic: a review of qualitative studies on the progression from initial use to abuse. Dialogues Clin Neurosci. 2017;19(3):259–269. doi: 10.31887/DCNS.2017.19.3/tcicero. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Howell L.L., Cunningham K.A. Serotonin 5-HT2 receptor interactions with dopamine function: implications for therapeutics in cocaine use disorder. Pharmacol Rev. 2015;67(1):176–197. doi: 10.1124/pr.114.009514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Venton B.J., Seipel A.T., Phillips P.E., Wetsel W.C., Gitler D., Greengard P., et al. Cocaine increases dopamine release by mobilization of a synapsin-dependent reserve pool. J Neurosci. 2006;26(12):3206–3209. doi: 10.1523/JNEUROSCI.4901-04.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Solinas M., Belujon P., Fernagut P.O., Jaber M., Thiriet N. Dopamine and addiction: what have we learned from 40 years of research. J Neural Transm. 2019;126(4):481–516. doi: 10.1007/s00702-018-1957-2. [DOI] [PubMed] [Google Scholar]
  • 250.George B.E., Dawes M.H., Peck E.G., Jones S.R. Altered accumbal dopamine terminal dynamics following chronic heroin self-administration. Int J Mol Sci. 2022;23(15) doi: 10.3390/ijms23158106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Jalabert M., Bourdy R., Courtin J., Veinante P., Manzoni O.J., Barrot M., et al. Neuronal circuits underlying acute morphine action on dopamine neurons. Proc Natl Acad Sci U S A. 2011;108(39):16446–16450. doi: 10.1073/pnas.1105418108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Wang L., Gao M., Wang Q., Sun L., Younus M., Ma S., et al. Cocaine induces locomotor sensitization through a dopamine-dependent VTA-mPFC-FrA cortico-cortical pathway in male mice. Nat Commun. 2023;14(1):1568. doi: 10.1038/s41467-023-37045-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Alex K.D., Pehek E.A. Pharmacologic mechanisms of serotonergic regulation of dopamine neurotransmission. Pharmacol Ther. 2007;113(2):296–320. doi: 10.1016/j.pharmthera.2006.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Navailles S., De Deurwaerdere P. Presynaptic control of serotonin on striatal dopamine function. Psychopharmacology (Berl). 2011;213(2-3):213–242. doi: 10.1007/s00213-010-2029-y. [DOI] [PubMed] [Google Scholar]
  • 255.Olson D.E. Psychoplastogens: a promising class of plasticity-promoting neurotherapeutics. J Exp Neurosci. 2018;12 doi: 10.1177/1179069518800508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Cameron L.P., Tombari R.J., Lu J., Pell A.J., Hurley Z.Q., Ehinger Y., et al. A non-hallucinogenic psychedelic analogue with therapeutic potential. Nature. 2021;589(7842):474–479. doi: 10.1038/s41586-020-3008-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Cohen-Cory S., Kidane A.H., Shirkey N.J., Marshak S. Brain-derived neurotrophic factor and the development of structural neuronal connectivity. Dev Neurobiol. 2010;70(5):271–288. doi: 10.1002/dneu.20774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Majić T., Schmidt T.T., Gallinat J. Peak experiences and the afterglow phenomenon: when and how do therapeutic effects of hallucinogens depend on psychedelic experiences? J Psychopharmacol. 2015;29(3):241–253. doi: 10.1177/0269881114568040. [DOI] [PubMed] [Google Scholar]
  • 259.Madsen M.K., Fisher P.M., Burmester D., Dyssegaard A., Stenbaek D.S., Kristiansen S., et al. Psychedelic effects of psilocybin correlate with serotonin 2A receptor occupancy and plasma psilocin levels. Neuropsychopharmacology. 2019;44(7):1328–1334. doi: 10.1038/s41386-019-0324-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Peng Y., McCorvy J.D., Harpsoe K., Lansu K., Yuan S., Popov P., et al. 5-HT(2C) receptor structures reveal the structural basis of GPCR polypharmacology. Cell. 2018;172(4):719–30 e14. doi: 10.1016/j.cell.2018.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Ubhayarathna M., Langmead C.J., Diepenhorst N.A., Stewart G.D. Molecular and structural insights into the 5-HT(2C) receptor as a therapeutic target for substance use disorders. Br J Pharmacol. 2024;181(22):4414–4429. doi: 10.1111/bph.16233. [DOI] [PubMed] [Google Scholar]
  • 262.Jastrzebska J., Frankowska M., Smaga I., Hubalewska-Mazgaj M., Suder A., Pieniazek R., et al. Evaluation of the 5-HT(2C) receptor drugs RO 60-0175, WAY 161503 and mirtazepine in a preclinical model of comorbidity of depression and cocaine addiction. Pharmacol Rep. 2023;75(1):99–118. doi: 10.1007/s43440-022-00428-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Zhang G., Wu X., Zhang Y.M., Liu H., Jiang Q., Pang G., et al. Activation of serotonin 5-HT(2C) receptor suppresses behavioral sensitization and naloxone-precipitated withdrawal symptoms in morphine-dependent mice. Neuropharmacology. 2016;101:246–254. doi: 10.1016/j.neuropharm.2015.09.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Lanteri C., Salomon L., Torrens Y., Glowinski J., Tassin J.P. Drugs of abuse specifically sensitize noradrenergic and serotonergic neurons via a non-dopaminergic mechanism. Neuropsychopharmacology. 2008;33(7):1724–1734. doi: 10.1038/sj.npp.1301548. [DOI] [PubMed] [Google Scholar]
  • 265.Willins D.L., Meltzer H.Y. Serotonin 5-HT2C agonists selectively inhibit morphine-induced dopamine efflux in the nucleus accumbens. Brain Res. 1998;781(1-2):291–299. doi: 10.1016/s0006-8993(97)01267-5. [DOI] [PubMed] [Google Scholar]
  • 266.Wu X., Pang G., Zhang Y.M., Li G., Xu S., Dong L., et al. Activation of serotonin 5-HT(2C) receptor suppresses behavioral sensitization and naloxone-precipitated withdrawal symptoms in heroin-treated mice. Neurosci Lett. 2015;607:23–28. doi: 10.1016/j.neulet.2015.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Brandt L., Jones J.D., Martinez S., Manubay J.M., Mogali S., Ramey T., et al. Effects of lorcaserin on oxycodone self-administration and subjective responses in participants with opioid use disorder. Drug Alcohol Depend. 2020;208 doi: 10.1016/j.drugalcdep.2020.107859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Levin F.R., Mariani J.J., Pavlicova M., Choi C.J., Basaraba C., Mahony A.L., et al. Lorcaserin treatment for extended-release naltrexone induction and retention for opioid use disorder individuals: a pilot, placebo-controlled randomized trial. Drug Alcohol Depend. 2021;219 doi: 10.1016/j.drugalcdep.2020.108482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Brezing C.A., Sibai M., Choi C.J., Mitra S., Mariani J.J., Naqvi N., et al. Open label pilot of lorcaserin (a serotonin 2C-receptor agonist) for cannabis use disorder. J Subst Use. 2024;29(4):487–494. doi: 10.1080/14659891.2023.2202760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Arout C.A., Cooper Z.D., Reed S.C., Foltin R.W., Comer S.D., Levin F.R., et al. 5HT-2C agonist lorcaserin decreases cannabis self-administration in daily cannabis smokers. Addict Biol. 2021;26(4) doi: 10.1111/adb.12993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Urbina F., Lowden C.T., Culberson J.C., MegaSyn Ekins S. Integrating generative molecular design, automated analog designer, and synthetic viability prediction. ACS Omega. 2022;7(22):18699–18713. doi: 10.1021/acsomega.2c01404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Kearney S.E., Zahoránszky-Kőhalmi G., Brimacombe K.R., Henderson M.J., Lynch C., Zhao T., et al. Canvass: a crowd-sourced, natural-product screening library for exploring biological space. ACS Cent Sci. 2018;4(12):1727–1741. doi: 10.1021/acscentsci.8b00747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Ekins S. CRC Press; 2025. An introduction to generative Drug Discovery. [Google Scholar]
  • 274.Ekins S., Williams A.J., Krasowski M.D., Freundlich J.S. In silico repositioning of approved drugs for rare and neglected diseases. Drug Discov Today. 2011;16:298–310. doi: 10.1016/j.drudis.2011.02.016. [DOI] [PubMed] [Google Scholar]
  • 275.Alves V.M., Korn D., Pervitsky V., Thieme A., Capuzzi S.J., Baker N., et al. Knowledge-based approaches to drug discovery for rare diseases. Drug Discov Today. 2022;27(2):490–502. doi: 10.1016/j.drudis.2021.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Puhl A.C., Raman R., Havener T.M., Minerali E., Hickey A.J., Ekins S. Identification of new modulators and inhibitors of palmitoyl-protein thioesterase 1 for CLN1 batten disease and cancer. ACS Omega. 2024;9(10):11870–11882. doi: 10.1021/acsomega.3c09607. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Neurotherapeutics are provided here courtesy of Elsevier

RESOURCES