Abstract
Herein we detail the first disclosure of VU0467319 (VU319), an M1 Positive Allosteric Modulator (PAM) clinical candidate that successfully completed a Phase I Single Ascending Dose (SAD) clinical trial. VU319 (16) is a moderately potent M1 PAM (M1 PAM EC50 = 492 nM ± 2.9 nM, 71.3 ± 9.9% ACh Max), with minimal M1 agonism (EC50 > 30 μM), that displayed high CNS penetration (Kps > 0.67 and Kp,uus > 0.9) and multispecies pharmacokinetics permissive of further development. Based on robust efficacy in multiple preclinical models of cognition, an ancillary pharmacology profile devoid of appreciable off-target activities, and a lack of cholinergic adverse effects (AEs) in rats, dogs and nonhuman primates, VU319 advanced into IND-enabling studies. After completing 4-week rat and dog GLP toxicology without AEs, including absence of cholinergic effects, the first in human Phase I SAD clinical trial of VU319 (NCT03220295) was performed at Vanderbilt, where a similar lack of adverse effects, including absence of cholinergic effects was noted. Moreover, signals of target engagement were seen at the highest dose tested. Thus, VU319 demonstrated the feasibility of achieving selective targeting of central M1 muscarinic receptors without eliciting cholinergic AEs that have plagued other drugs targeting CNS cholinergic neurotransmission.
Keywords: muscarinic acetylcholine receptor subtype 1 (M1), positive allosteric modulator (PAM), cognition, metabolism
Introduction
The selective activation of muscarinic acetylcholine receptors (mAChRs or M1–5) has been a long sought after goal since clinical trials in the 1980s demonstrated that “M1 agonists” had robust efficacy in improving cognition in Alzheimer’s patients; however, these unselective mAChR agonists activated peripheral M2 and M3 receptors resulting in significant adverse cholinergic events, SLUDGE (salivation, lacrimation, urination, defecation, gastrointestinal distress and emesis).1−6 The most successful of these unselective agonists was xanomeline (1), an M1/M4 preferring agonist (Figure 1), that at therapeutic concentrations, activates all the mAChRs. Karuna employed a creative strategy to revive 1 recently, gaining FDA approval of the combination of xanomeline 1 coadministered with the peripherally restricted muscarinic antagonist trospium to reduce SLUDGE effects (combination known as KarXT, cobenfy).7−9 While cobenfy represents an important advance, balancing the degree of mAChR activation and inhibition can lead to adverse effects across heterogeneous patient populations. As a chronic, daily maintenance therapeutic, a highly selective M1 activator, devoid of the need for coadministration of a peripheral mAChR antagonist may offer a more attractive option for many patients.10
Figure 1.

Structures of the M1/M4-preferring xanomeline (1), representative M1 PAMs with cholinergic AEs 2-5, and representative M1 PAMs with lesser levels of M1 agonism 6-10, that showed no cholinergic AEs.
Over 20 years ago, the concept of allosteric modulation of GPCRs was exploited to provide highly selective positive allosteric modulators (PAMs) of both Family A and C GPCRs,11,12 including the first M1 PAM, BQCA (2) by Merck.13−15 Since those initial discoveries, our laboratories and others have been developing M1 PAMs with a diverse array of pharmacological profiles (Figure 1). Clinical data, genetic data and preclinical data with tools 1-10 strongly suggests M1 PAMs can treat cognitive dysfunction in schizophrenia, Alzheimer’s disease, prion diseases and other CNS disorders.14−26 However, caveats appeared and demonstrated that all M1 PAMs are not equivalent; moreover, intrinsic agonism, signal bias and the degree of cooperativity may have been key factors in adverse events, cholinergic side effects and candidate advancement into the clinic.14−26 Based on these caveats, and data reported with M1 PAMs from prior programs, we took steps to derisk our M1 PAM chemotypes, both preclinically and clinically.
Here, we detail for the first time the discovery and development of VU319, a novel M1 PAM, minimal agonism, that displayed robust pro-cognitive efficacy in preclinical models and was devoid of SLUDGE effects in mice, rats, dogs and nonhuman primates. In a Phase I SAD clinical trial of VU319 (NCT03220295),27−30 SLUDGE effects were not observed, and signs of precognitive efficacy were evident. Herein, we describe the discovery of VU319, its in vitro and in vivo pharmacological and DMPK profile, its safety/toxicology in IND-enabling studies and its progression into human testing.
Results and Discussion
Design
Results with BQCA (2), a potent M1 PAM with high intrinsic M1 agonist activity, indicated that this pharmacological profile may be undesirable, and that an M1 PAM with lower levels of intrinsic M1 agonism might be better tolerated an cause fewer cholinergic adverse evenets.13−15,17 A functional M1 high-throughput screen at Vanderbilt identified BQCA-like PAMs, and a single chemotype that displayed no M1 agonism up to 30 μM, a simple isatin derivative, VU0119498 (11).31 However, 11 proved to be a PAM of all three Gq-coupled mAChRs (M1, M3 and M5) but inactive on the Gi/o-coupled mAChRs (M2 and M4); moreover, 11 harbored an electrophilic and reactive isatin moiety, rendering it unattractive as a lead for an M1 PAM development effort.31 However, with only a single hit lacking appreciable M1 agonism, the medicinal chemists had no choice but to attempt to remove the undesirable isatin moiety of 11 while developing SAR to engender selective M1 PAM pharmacology with minimal M1 agonism. As previously described, optimization efforts on the southern benzyl tail identified modifications that abolished M3 and M5 PAM activity, affording a selective M1 PAM, ML137 (12), with minimal agonism, yet still possessing the isatin core.32 Deletion of the ketone moiety in 12 afforded lactam 13 (VU0448350), a weak but selective M1 PAM, again with minimal M1 agonism.33 Surveying alternative positional lactam isomers led to the discovery of VU0451725 (14), a selective M1 PAM with minimal agonism. An aza scan of the phenyl moiety then afforded VU0453595 (15), a selective M1 PAM that found utility as a rodent tool compound for selective M1 potentiation, again without M1 agonism.34 Finally, extensive chemical optimization of the southern benzyl tail region led to the discovery of VU319 (16) (Figure 2).
Figure 2.
Roadmap to the discovery of VU0467319 (VU319, 16). From an unselective, pan-Gq-mAChR (M1, M3, M5) PAM 11, harboring an electrophilic and reactive isatin moiety, to a highly selective M1 PAM clinical candidate, VU319 (16) with minimal agonism and no reactive functionality.
Discovery and Process Chemistry
For the synthesis of VU319 we developed two synthetic routes: (1) the optimized medicinal chemistry route and (2) the process route employed in the CMC tox lot and GMP supply campaigns.35 The optimized discovery chemistry route to VU319 was a convergent route employing commercially available starting materials (Scheme 1). Staring with indazole boronic ester 17, a Suzuki coupling with bromobenzaldehyde 18 proceeded smoothly, followed by conversion to the oxime and Zn-mediated reduction to key benzylamine 19 in 79% overall yield for the three-step sequence. In parallel, commercial nicotinic ester 20 was chlorinated with trichloroisocyanuric acid (TCCA) to deliver 20 in quantitative yield. Finally, amine 19 was reacted with benzyl chloride 21 in the presence of Hünig’s base at 80 °C leading to lactam formation and the production of VU319 in 40% yield. Overall, the convergent discovery synthesis of VU319 proceeded in five steps with an overall yield of 31.6%.35 The CMC process route for a 1.3 kg tox lot and GMP manufacture of a 3.3 kg lot employed a similar route, but key reactions were changed to avoid potential genotoxic intermediates (Scheme 2). Boronic acid 17, while cost-effective in the discovery chemistry campaign, proved too costly, and difficult to obtain in the necessary quantities, to employ directly as a starting material in the CMC campaign. Thus, commercial bromoindazole 22 was designated starting material 1 (SM 1) and converted into 17 under standard conditions in excellent yield. The first GMP step proceeded with the Suzuki coupling of 17 with 18 (starting material 2, SM 2) under the discovery chemistry conditions to deliver aldehyde 23. The second GMP step was conversion to the oxime to provide intermediate 3 (24), followed by step 4, the Zn-mediated reduction to the benzyl amine 19 (step 3). The benzyl chloride intermediate 21 in the discovery route was flagged as a potential genotoxic intermediate, so the team revised the final coupling steps. Ultimately, commercial nicotinic ester 20 (starting material 3, SM 3) was oxidized with selenium dioxide to the corresponding aldehyde 25. A reductive amination protocol with 19 and 25 employing STAB (step 4) produced VU319 on scales up to 3.3 kg under cGMP as a white solid. Crude VU319 was recrystallized from ethanol at 60–65 °C. Product was acquired over only four GMP steps in 99.7% purity and with individual impurities ≤0.10% and an overall yield of 50.5% (3.3 kg).35 VU319, 6-(2,6-difluoro-4-(2-methyl-2H-indazol-4-yl)benzyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-5-one, had a clean, sharp melt at 208 °C, a molecular weight of 390.4, an experimental logD7.4 of 3.28 and a pKa of 1.4.
Scheme 1. Discovery Chemistry Route for the Synthesis of VU319 (16).
Reagents and conditions: (a) PdCl2(dppf)·DCM, Cs2CO3, THF:H2O, 40 °C, 12 h; (b) NH2OH·HCl, CH3COONa, EtOH, 2.5 h; (c) Zn, acetic acid, 2.5 h, 79% over three steps; (d) TCCA, DCM, rt, 24 h, 100%; (e) Hünig’s base, CH3CN, 80 °C, 12 h, 40%.
Scheme 2. CMC Process Route for the Synthesis of VU319 (16) on 3.3 kg Scale.
Reagents and conditions: (a) Bis(pinacolato)diboron, PdCl2(dppf)·DCM, 1,4-dioxane, KOAc, 85–90 °C, 10 h, 66%; (b) PdCl2(dppf)·DCM, Cs2CO3THF:H2O, 55–60 °C, 3 h, 95%; (c) NH2OH·HCl, CH3COONa, EtOH, 5 h, 95%; (d) Zn, acetic acid,2.5 h, 66%; (e) SeO2, 1,4-dioxane, 135–140 °C, 2 h, 51%; (f) STAB, ZnCl2, THF, 50–55 °C, 6 h, 73%.
Molecular Pharmacology
After two decades of research with M1 allosteric agonists,36−38 and M1 PAMs with minimal to substantial levels of agonism,15,17,20−22,26,31−34 we felt that a translatable candidate had to avoid overstimulation of the M1 receptor, in order to prevent cholinergic side effects and diminished efficacy at high doses and/or with chronic administration. Our team determined that we needed to develop an M1 PAM with minimal agonism in both our stably transfected, TET-inducible mAChR cell lines as well as in native tissues (induction of long-term depression, LTD).15,17,20−22,26,31−34 Moreover, the vast majority of M1 PAMs that were advanced into clinical testing were very potent, but with low Kps/Kp,uus, typically less than 0.1, leading to overstimulation of the M1 receptor (particularly peripheral M1 receptors), Racine scale 4/5 seizures in mice, robust induction of LTD in native tissues, cholinergic side effects and diminished pro-cognitive efficacy.17,19 Moreover, when we began investigating allosteric modulation of GPCRs, we arbitrarily selected an EC20 concentration of acetylcholine as the subthreshold level of endogenous agonist for which to assess PAM activity (i.e., we desired a large PAM window).31 In vivo, cholinergic tone/ACh levels vary across brain regions, in different disease states and numerous other caveats; therefore, it is possible that the in vivo potency of our M1 PAMs could have been underestimated by the cell-based functional assays.17 As a result, our strategy was to translate a moderately potent M1 PAM in our cell-based assay, with minimal M1 agonism, good CNS penetration and overall drug-like properties to potentiate whatever ACh tone is present in vivo. VU319 delivered on this unique and desired profile.
Figure 3A highlights the human M1 PAM concentration–response-curve (CRC) in the presence of an ∼ EC20 of ACh (M1 PAM EC50 = 492 nM ± 2.92 nM, 71.3 ± 9.9% ACh Max, n = 94) as well as the effect on M1 receptor activation in the absence of ACh (ie., the M1 agonist CRC).35 At a concentration of 30 μM, there is only slight activation, highlighting that VU319 is a PAM with minimal M1 agonism (M1 agonism >30 μM). As allosteric sites may not be conserved across species, we evaluated the ability of VU319 to potentiate mouse, rat, and cynomolgus monkey (cyno) M1 (Figure 3B) to enable translation. Here, VU319 potentiated M1 in all three species: rat (M1 EC50 = 398 ± 195 nM, 81.3 ± 11.3% ACh Max, n = 13), mouse (M1 EC50 = 728 ± 184 nM, 55.9 ± 5.6% ACh Max, n = 2 in triplicate) and cyno (M1 EC50 = 374 nM, 57.8% ACh max, n = 1 individual experiment performed in duplicate). Moreover, PAM 16 was selective (EC50 > 30 μM) versus M2–5 for both human (Figure 4A) and rat (Figure 4B).35
Figure 3.
(A) VU319 is a PAM of the human M1 muscarinic receptor with minimal M1 agonism. Data represent one experiment performed in triplicate. (B) VU319 is a PAM of the rat, mouse, and cynomolgus monkey M1 muscarinic receptors. Representative data from one experiment for each species performed in triplicate.
Figure 4.
VU319 is highly selective as an M1mAChR PAM at human (A) and rat (B) M1–M5 receptors. Increasing concentrations of VU319 were applied approximately 2 min prior to an appropriate concentration of acetylcholine (ACh) designed to elicit an EC20 response. Calcium mobilization responses were measured and normalized to a maximal ACh response elicited after activation of each receptor.
In a competitive radioligand binding assay, VU319 did not displace orthosteric [3H] N-methylscopolamine (NMS) binding at the muscarinic receptors (up to 30 μM), indicating an allosteric mechanism of action (Figure 5A).35 In contrast, the orthosteric antagonist atropine displaced [3H]-NMS binding with a Ki of 1.8 nM, in accord with literature values. As shown in Figure 5B, VU319 shifted the ACh concentration–response curve to the left in a calcium mobilization assay in cells expressing the rat M1 receptor, with a maximum leftward shift of 96-fold at 30 μM (and 38-fold at 10 μM). PAMs act via increasing the affinity (α cooperativity factor), efficacy (β cooperativity factor), or both of an endogenous orthosteric agonist such as ACh. Here, increasing concentrations of 16 progressively shifted the ACh displacement curve to the left in CHO cells expressing the rat M1 receptor. This resulted in a calculated α value of 59, (see Supporting Information) suggesting that part of the mechanism of receptor potentiation induced by VU319 is via an increase in ACh affinity at M1. Moreover, and in contrast to the results observed at M1, VU319 was ineffective in shifting ACh affinity at rat M2, M3, M4 or M5 receptors. An early exploration of signal bias discovered that VU319 also potentiated the recruitment of β-arrestin2 to the human M1 receptor in a concentration dependent manner (EC50 = 890 nM, 72% max).35 Finally, we assessed probe dependence of VU319 and its ability to potentiate other orthosteric agonists. In addition to ACh, VU319 also potentiated the response of oxotremorine-M (M1 EC50 = 400 nM, 59% ACh Max, M2–5 EC50 > 30 μM), but was ineffective at potentiating xanomeline, clozapine or N-desmethylclozapine (NDMC).35
Figure 5.
(A) In contrast to atropine, VU319 does not displace the binding of the orthosteric radioligand, [3H] NMS. (B) Fold shift of the ACh concentration–response curve in a calcium assay in cells expressing the rat M1 receptor and exposed to increasing concentrations of VU319 prior to a full range of ACh concentrations. Data represent two experiments performed in duplicate or triplicate.
Drug Metabolism and Pharmacokinetics
VU319 displayed a very attractive in vitro and in vivo DMPK profile, and for the time (ca. 2016–2017) demonstrated improved CNS penetration relative to other M1 PAMs reported.35 In rodents, VU319 displayed good unbound fraction in both plasma (fu (rat, mouse) = 0.028, 0.028) as well as brain homogenate binding (BHB fu (rat, mouse) = 0.040, 0.048). Similarly, human (plasma fu = 0.034), dog (plasma fu = 0.076) and cyno (plasma fu = 0.059) all displayed acceptable plasma protein binding. Moreover, VU319 appeared stable in vitro, with low predicted hepatic clearance values across species (CLhep (human, rat, dog cyno): 1.3, 5.7, 5.5, 5.6 mL/min/kg) and an acceptable CYP450 inhibition profile (3A4, 2B6, 2D6: IC50 > 30 μM; 1A2: IC50 = 14 μM; 2C9: IC50 = 5.1 μM). Follow-up studies showed no significant CYP450 (3A4, 2B6 and 1A2) induction liability, and CYP450 phenotyping indicated CYP3A4 solely contributed to the metabolism of VU319. In terms of predicted CNS penetration in man, VU319 was not a human P-gp substrate (MDCK-MDR1 ER = 1.6) or MDCK-BCRP substrate (ER = 1.8) with high permeability (Papp = 31 × 10–6 cm/s). In the FDA transporter panel, no issues arose, including BSEP (IC50 > 30 μM). Mini-AMES was negative in two strains (TA98 and TA100 with and without S9), and GSH trapping studies in microsomes across species (h, r, c, d) found only a trace (<0.1%) of adducts.35 These positive data paved the way to initiate in vivo PK studies.
In vivo, VU319 displayed significantly better brain exposure in rodents than other PAMs of the time (plasma:brain partitioning Kps < 0.1; Kp,uus ≪0.1). In mouse, VU319 provided a Kp of 0.77 and a Kp,uu of 1.3, while in rat, VU319 displayed a Kp of 0.64 and a Kp,uu of 0.91.35 M1 PAMs at the time were very potent, but had low Kp values, leading to relatively low CNS exposures and cholinergic AEs.17 The goal was to provide higher CNS exposure, but with moderate M1 PAM potency to effectively potentiate varying ACh tone across brain regions.
Multispecies IV/PO PK studies further supported the low in vitro predicted hepatic clearance (Table 1). Across mouse, rat, dog and cyno, there was a robust in vitro:in vivo correlation (IVIVC) with VU319 showing low clearance (Clp (m, r, d, c): 25.4, 3.0, 4.0, and 3.3 mL/min/kg), low to moderate volumes (Vss 0.67–2.2 L/kg) and attractive half-lives (t1/2 (m, r, d, c): 4.1, 3.0. 7.5 and 4.3 h). Despite modest solubility (FaSSIF, 7 μM and SGF, 36 μM), VU319 was readily absorbed across species affording early Tmax (1–2 h) and with excellent oral bioavailability (%F (m, r, d, c): 80, 93, 100, and 59).35 Thus, VU319 possessed an attractive profile for continued derisking and development toward a clinical candidate.
Table 1. IV/PO Pharmacokinetic Parameters of 16.
| parameter | mouse | rat (SD) | dog (beagle) | NHP (cyno) |
|---|---|---|---|---|
| dose (mg/kg) iv/po | 1/3 | 1/3 | 1/3 | 1/3 |
| CLp (mL/min/kg) | 25.4 | 3.0 | 4.0 | 3.3 |
| Vss (L/kg) | 2.2 | 0.67 | 2.1 | 0.90 |
| elimination t1/2 (h) | 4.1 | 3.0 | 7.5 | 4.3 |
| F (%) po | 80 | 93 | 100 | 59 |
| Cmax (μM) | 3.1 | 4.5 | 3.3 | 3.7 |
| Tmax (h) | 1 | 1 | 2 | 2 |
| AUC (μM*h) | 39 | 43 | 47 | 23 |
| Kp | 0.77 | 0.64 | ||
| Kp,uu | 1.3 | 0.91 |
Derisking Cholinergic AEs
In our optimization work-flow, we incorporate a high-throughput phenotypic seizure liability assay (100 mg/kg intraperitoneal (i.p.)), as mice are very sensitive to cholinergic mechanisms and they readily display Racine scale seizures when the M1 receptor is over stimulated.26,39,40 This is a high-bar assay, as this sets a very conservative exposure for initiation of cholinergic AEs. Here (Figure 6), a high dose (100 mg/kg intraperitoneal (IP)) of BQCA (2), a potent M1 PAM significant M1 agonism,13−15 rapidly initiated Racine scale 3/4 seizures 30 min post administration. In contrast, VU319 did not induce seizure liability up to 6 h post administration, consistent with other M1 PAMs, with minimal M1 agonism. A satellite IP PK study (100 mg/kg IP in C57Bl6 mice, 30% captisol) demonstrated that total brain exposures were 85.4 μM (117-fold above the mouse M1 EC50).35 As discussed in the past, pharmacodynamic effects for dozens of M1 PAMs correlates more closely with total brain than with free brain concentrations, as the arbitrarily selected EC20 value for the PAM EC50 likely underestimates endogenous ACh tone. To assess peripheral cholinergic toxicity, we performed a modified Irwin neurological battery to score autonomic or somatosensory side effects (Figure 7). At a dose of 56.6 mg/kg IP, BQCA (2) produced significant SLUDGE effects in mice. In contrast, VU319 at doses of either 56.6 mg/kg IP (60.4 μM, 83-fold above the mouse M1 PAM EC50) or at 100 mg/kg IP (85 μM, ∼ 117-fold above the mouse M1 EC50), had no SLUDGE noted.
Figure 6.

Racine Score test in mice. Pretreatment with M1 PAMs (100 mg/kg, i.p., 10 mL/kg, 180 min) BQCA (2) resulted in robust behavioral convulsions at 30 min post administration, while VU319 did not cause any observed adverse effects out to 6 h post administration. N = 3/group of male C57Bl/6 mice. ANOVA p < 0.0001; ****p < 0.0001 as compared to vehicle control.
Figure 7.
Effects of VU319 on the modified Irwin Neurological Test battery in mice. At doses of either 56.6 mg/kg or 100 mg/kg IP, and over a 6 h time course, no adverse cholinergic events (SLUDGE) were noted.
In a Eurofins Lead Profiling radioligand panel of 68 GPCRs, ion channels and transporters tested at 10 μM, VU319 only displaced radioligand binding at two (α2a, Ki = 2.7 μM and imidazoline I2 central, Ki = 2.8 μM) of the 68 targets. In follow-up functional assays, VU319 was inactive at both targets.35 Moreover, VU319 did not displace hERG radioligands, but in a functional EP assay, VU319 displayed an IC50 of 12 μM (a value we could go forward with).
A hallmark of potent M1 PAMs with intrinsic M1 agonism is M1 receptor overstimulation and induction of Long-Term Depression (LTD) in layer V medial prefrontal cortex (mPFC).26 Therefore, we wanted to explore if VU319 would be devoid of LTD-induction. In electrophysiological studies in native mouse tissues, VU319 did not induce any significant change in field excitatory post synaptic potentials (fEPSPs) recorded from layer V and evoked by electrical stimulation in layer II/III at either 10 μM (∼14× above the functional mouse EC50), or 30 μM concentrations (∼42× above the functional mouse EC50). Thus, VU319 maintains activity dependence of PFC function, is devoid of seizure liability and does not show induce SLUDGE, in mice up to 100 mg/kg, indicating the absence of cholinergic toxicity in the most sensitive species to observe these adverse effects. Moreover, the pharmacological profile of VU319 clearly differentiates from the M1 PAMs with high intrinsic M1 agonism that dominated the field at the time (2-5, 2009–2016).13−26
In Vivo Behavior
VU319 possessed a unique profile of modest M1 PAM potency (with minimal M1 agonism) while displaying CNS penetration 6–10-fold better than prior M1 PAMs 2-5.13−26 While the profile of VU319 avoided cholinergic toxicity, would this profile translate to efficacy in preclinical cognition models? Historically, novel object recognition (NOR) has been our first-tier cognition assay to assess M1 PAM efficacy.26 In the study (Figure 8), pretreatment of VU319 (0.3 to 5.6 mg/kg PO) dose-dependently increased the recognition index during the NOR test in normal Sprague–Dawley rats (p = 0.0053, *p < 0.05) relative to the vehicle-treated control group (N = 10–18). The minimum effective dose (MED) was 1 mg/kg PO (1 μM total brain, ∼ 2.5× the rat M1 PAM EC50, Kp = 0.82), with a trend at 0.3 mg/kg (400 nM total brain, ∼ 1× the rat EC50).35 Maximum recognition index was achieved between 3 and 5.6 mg/kg (3.2 to 7.2 μM total brain, ∼ 7–16× the rat EC50). Thus, an M1 PAM of modest potency (rat EC50 = 398 nM, 81% ACh Max) with high CNS penetration proved to be efficacious at low doses.
Figure 8.

Effects of VU319 on Novel Object Recognition in rats. VU319 dose-dependently enhanced recognition memory in rats. Pretreatment with 0.3, 1, 3, and 5.6 mg/kg VU319 (p.o, 0.5% natrosol/0.015% Tween 80 in water, 30 min) prior to exposure to identical objects significantly enhanced recognition memory assessed 24 h later. N = 10–18/group of male Sprague–Dawley rats. ANOVA p = 0.0053, *p < 0.05.
There are multiple potential disease states that could potentially benefit from treatment with an M1 PAM. Previously we demonstrated the ability of PAM 7 to reverse cognitive deficits induced by the atypical antipsychotic risperidone,26 which is associated with cognitive symptom clusters in schizophrenia. Recent work from the Tobin lab suggests a key disease-modifying role in prion diseases,25 while the Niswender lab has shown efficacy in Rett syndrome.41 Beyond these disorders, multiple forms of dementia (mild cognitive impairment, vascular dementia, Lewey body dementia and Alzheimer’s disease (AD)) are potentially attractive therapeutic indications for an M1 PAM.1−6
Since acetylcholine esterase inhibitors (AChEIs) represent one of the only approved classes of drugs for the treatment of cognitive impairments observed in AD patients,42 it was important to also determine the potential additive effects of our M1 PAM when given in combination with an AChEI on cognitive functions. We assessed the in vivo efficacy of VU319 in combination with donepezil in the NOR task, a preclinical model of memory function in rats following a single oral administration. Male Sprague–Dawley rats were treated with VU319 formulated as a microsuspension in 20% (w/v) HPBCD in sterile water at concentrations of 0.03, 0.1, or 0.3 mg/mL and administered as a single 0.3, 1, or 3 mg/kg dose (10 mL/kg) by oral gavage. Rats were administered vehicle or VU319 60 min before a vehicle or 0.3 mg/kg intraperitoneal administration of donepezil (1 mL/kg). Data are expressed as mean ± SEM and were analyzed using a one-way ANOVA; if significant (p < 0.05), all dose groups were compared with the vehicle-treated group using a Dunnett’s post hoc test. As shown in Figure 9, the dose of 0.3 mg/kg donepezil alone did not significantly increase recognition memory relative to vehicle-treated controls; however, when given in combination with increasing doses of VU319, there was a dose-dependent enhancement of the recognition index during the NOR task (p = 0.0449, * p < 0.05) (N = 11–12). To ensure this synergy was not due to a drug–drug interaction (DDI), we preformed parallel PK studies to assess both agents as DDI perpetrators and victims. Fortunately, there was no DDI for either drug. Collectively, these data suggest that selective activation of M1 by VU319 may enhance the efficacy of AChEIs when given in combination in clinical populations.
Figure 9.

VU319 (PO), in combination with an inactive dose of donepezil (0.3 mg/kg IP) produced enhanced recognition memory in the NOR task in rats. Dose-dependent enhancement of the recognition index during the NOR task (p = 0.0449, one-way ANOVA with a dunnett post hoc test * p < 0.05) (N = 11–12).
Beyond these assays, VU319 dose-dependently reversed scopolamine-induced deficits in the acquisition of contextual fear conditioning (CFC), dose-dependently reversed scopolamine-induced deficits in the eight arm radial arm maze (RAM) task, dose-dependently reversed risperidone-induced deficits in CFC, increased active wake 1–2 h post dosing in EEG without impact on any other sleep parameters (consistent with cognitive promoting effects without disrupting sleep quality), but was inactive, as anticipated in reversing amphetamine-induced hyperlocomotion.35 Finally, we derisked other potential liabilities in vivo. VU319 has no disruptive effects on basal locomotor activity and motor coordination (rotorod) in rats at doses up to 10 mg/kg (10× above the MED in PD models).35
As we began to consider biomarker strategy, we knew PET would not work, as M1 affinity was low, but cooperativity was very high (leading to the nM functional PAM potency). Moreover, we were never able to discover and develop M1 Negative Allosteric Modulators (NAMs) to employ as radioligands and PET tracers competitive with VU319. Thus, the team explored possible functional biomarkers such as FMRI and EEG, and though not quantitative, these could potentially provide qualitative target engagement in Phase I. As shown in Figure 10, VU319 treatment decreased alpha and beta frequencies and increased both the low and high frequency gamma power in the frontal cortex, specifically when the rats were awake (p < 0.0001). However, VU319 treatment did not disrupt sleep quality as indicated by no change in delta power during SWS. During wake, pretreatment with PAM VU319 did not alter the power of either delta (p = 0.8910) or theta (p = 0.3495) frequencies at 1–2 h after dosing.35 Taken together, these data reveal that VU319 produces enhanced levels of arousal during wake, consistent with cognitive promoting effects, without disrupting sleep quality.
Figure 10.

VU319 (PO) produced increases in high gamma (γ) power in rats in a dose-dependent manner. For all dose–response studies a two-way analysis of variance was applied to examine effects of dose and frequency for each of the frequency bands; significance was defined as p < 0.05.
Metabolite Identification
With enthusiasm for VU319 as a candidate, there remained a few additional sets of data needed to confirm its candidacy. First and foremost, we needed to ensure that we had metabolite coverage across our IND-toxicology safety species and that there were no unique human metabolites. Preliminary biotransformation work in multispecies liver S9 microsomes was later confirmed with VU319 incubations in human (H), rat (R), dog (D) and monkey (P, primate) hepatocytes. A total of seven metabolites were identified in these studies (Figure 11). VU319 was the major component in human (65%), rat (76%) and dog (71%), but lower in monkey (25%) by UV absorbance peak area. In all species, Metabolite D (VU0481424, 26) was the most abundant metabolite: human (34%), rat (23%), dog (28%) and monkey (74%). The other metabolites were of very low abundance (<0.3%). Importantly, there were unique human metabolites. As VU0481424 (Metabolite D, 26) was produced in significant quantities, we had to synthesize it (Scheme 3) and assess its pharmacology and DMPK profiles. The synthesis of 26 proved straightforward. Starting from commercial anhydride 27, treatment with benzyl amine 19 and triethylamine followed by HATU coupling conditions afford the succinimide congener 28 in 90% yield. Several reductants for the chemoselective reduction of 28 to afford 26 were evaluated; ultimately, NaBH4, possibly via chelation to the pyridine nitrogen, provided 26 in 24% yield.35
Figure 11.
Multispecies hepatocyte metabolite identification of VU319. Metabolite D (VU0481424, 26) is a major, inactive metabolite.
Scheme 3. Synthesis of Metabolite D (VU0481424, 26).
Reagents and conditions: (a) 19, Et3N, DMF/MeCN, rt, 1 h, then HATU, 90%; (b) NaBH4, DCM/MeOH, 24%.
Metabolite D (26) proved to be an inactive metabolite (human M1–5 EC50s > 10 μM); however, it proved to have a good DMPK profile, despite having an acylated aminal functional group. Like the parent VU319, 26 displayed good unbound fraction in both rat plasma (fu = 0.038) as well as brain homogenate binding (BHB fu = 0.046). Similarly, human (plasma fu = 0.063), dog (plasma fu = 0.12) and cyno (plasma fu = 0.06) all displayed acceptable plasma protein binding. Moreover, 26 appeared stable in vitro, with low predicted hepatic clearance values across species (CLhep (human, rat, dog cyno): 11.2, 34.5, 11.7, 16 mL/min/kg) and an acceptable CYP450 inhibition profile (3A4, 2B6, 2D6: IC50 > 30 μM; 1A2: IC50 = 0.19 μM; 2C9: IC50 = 13 μM), except for 1A2. The metabolite 26 was also brain penetrant, with a rat Kp of 0.29 and a Kp,uu of 0.35, and an excellent rat PK profile (Clp = 7.9 mL/min/kg, Vss= 1.4 L/kg, t1/2 = 4 h). Ancillary pharmacology profiling was clean, so there were no pharmacological concerns with the presence of 26, but we would have to monitor it in the 28-day IND toxicology and in the Phase I SAD clinical trial.
The last remaining data needed were human dose projections, to determine if both development and cost of goods for VU319 was warranted. With the in vitro and in vivo multispecies DMPK data for VU319 in hand, and exposures/effective concentrations of VU319 from the NOR assay, we evaluated human PK parameters and dose projections using six different methods (Table 2). The average of all six methods predicted VU319 to be a low clearance compound in man (CLp < 1.3 mL/min/kg)) with a reasonable volume (Vss 0.52–1.2 L/kg) and a long half-life (up to 12 h). Projected human doses ranged from 150 to 440 mg for 24 h QD coverage to 50–100 mg for 24 h BID. A more likely scenario would be 12–16 h coverage, where projected QD doses range from 75 to 195 mg, or BID dose range from 40 to 70 mg. As these are not steady state PK approximations, dose projections could improve based on actual human PK in Phase I. Moreover, if given in combination with donepezil, it theoretically might be possible to use a lower dose of VU319.
Table 2. Human PK and Efficacious Dose Projections for VU319.
| compound (ID) | prediction method | predicted human PK parameter | projected human efficacious dose (mg)a | |||||||
|---|---|---|---|---|---|---|---|---|---|---|
| 24 h coverageb | 16 h coveragec | 12 h coveraged | ||||||||
| CLp (mL/min/kg) | Vss (L/kg) | t1/2 (h) | QD (24 h tau) | BID (12 h tau) | QD (24 h tau) | BID (8 h tau) | QD (24 h tau) | BID (6 h tau) | ||
| VU0467319 | HLMe | 1.3 | 0.76 | 6.8 | 440 | 100 | 195 | 70 | 135 | 65 |
| HLM R/D/C IVIVEf | 0.79 | 0.76 | 11 | 150 | 50 | 90 | 40 | 75 | 40 | |
| SSS ratg | 0.93 | 0.81 | 10 | 195 | 60 | 115 | 50 | 85 | 45 | |
| SSS dogg | 1.1 | 0.94 | 10 | 235 | 70 | 135 | 60 | 105 | 55 | |
| SSS cynog | 0.98 | 0.52 | 6.1 | 385 | 80 | 160 | 55 | 115 | 50 | |
| simple R/D/C allometryh | 1.1 | 1.2 | 12 | 190 | 65 | 125 | 60 | 100 | 55 | |
| all (range) | 0.79–1.1 | 0.52–1.2 | 6.1–12 | 150–440 | 50–100 | 90–195 | 40–70 | 75–135 | 40–65 | |
Mean Ka from rat and cyno oral PK studies (∼0.7/h, MAT approach) and oral F from cyno (0.59, lowest) used for all prediction methods (ref: Chiou et al. 1998; 2000 Pharm. Res. 15:11 and 17:2).
Doses projected to provide 24 h daily coverage of the targeted Cmin,p based on the Cmax,p from rat NOR model MED [1 mg/kg PO] corrected for species differences in fuplasma, and in vitro potency.
Doses projected to provide 16 h daily coverage of the targeted Cmin,p, based on the Cmax,p, from rat NOR model MED [1 mg/kg PO] corrected for species differences in fuplasma and in vitro potency.
Doses projected to provide 12 h daily coverage of the targeted Cmin,p, based on the Cmax,p from rat NOR model MED [1 mg/kg PO] corrected for species differences in fuplasma and in vitro potency.
Direct scaling of CL from human liver microsomes using the well-stirred model with fuplasma and calculated fumics predicted human Vss is mean from rat/dog/cyno SSS predictions.
Scaling of CLint from human liver microsomes according to the same method as described for “method a” but with correction for the mean fold-difference observed between in vitro and calculated in vivo unbound CLint in rat, dog, and cyno (approximately ∼0.6x); predicted human Vss, is mean from rat/dog/cyno SSS predictions.
Single-species allometric scaling of rat, dog, or cyno CLp, (exp. 0.75) and Vss (exp. = 1), respectively, corrected for species differences in fuplasma.
Multispecies allometric scaling of CLp and Vss (using empirically determined exponents) corrected for species differences in fuplasma.
Investigational New Drug (IND)-Enabling Studies
Based on the overall profile of VU319, and with a generous grant from the William K. Warren Foundation to support IND-enabling studies, we contracted with Davos Pharma for the CMC (Scheme 2), IND-enabling toxicology and regulatory services to draft the Investigational New Drug Application and take VU319 to an open IND. VU319 was negative in the standard IND battery of in vitro genotoxicity assays, no treatment-related findings were noted in respiratory rat, and no effect on any cardiac parameter was observed in CV dog. Dose-range finders (DRFs) and maximum tolerated dose (MTD) studies were conducted in male and female cohorts of rats, dogs and nonhuman primates, achieving exposures 10- to 20-fold over the rat NOR MED exposure (and >25-fold over the M1 PAM EC50) without any sign of SLUDGE or toxicity findings in the non-GLP histopathology analysis. GLP 28-day toxicology was performed in rat and dog, achieving exposures >20-fold the rat NOR MED without SLUDGE or observable AEs. The only clinical sign noted was mild weight loss, from which the animals fully recovered.
These data were compiled and submitted to the FDA as an Investigational New Drug (IND) Application on October 3, 2016, and the IND was opened on November 2, 2016. With the IND data package, and in collaboration with Dr. Paul Newhouse, the Director of the Center for Cognitive Medicine at Vanderbilt, a first-in-human Phase I single ascending dose (SAD) study with VU319 was initiated in the Vanderbilt Institute for Clinical and Translational Research (VICTR). NCT03220295 was initiated on July 18, 2018 (almost a decade after we discovered HTS hit 11).29 Subsequently, safety and pharmacokinetic data from that trial has been reported.27,28 In human volunteers, VU319 was well tolerated, with no cholinergic side effects (SLUDGE) noted, at doses showing signs of cognitive improvement and functional target engagement. A full account of the Phase I SAD VU319 is in preparation and will be published in due course.
Conclusions
In summary, a lead optimization campaign starting from a chemically reactive, nonselective M1,3,5 PAM, with minimal M1 agonism, was converted into VU319, a highly selective (>30 μM versus M2–5) and highly brain penetrant (rat Kp of 0.64, Kp,uu of 0.91) M1 PAM (EC50 = 492 nM, 71% ACh Max), while retaining the desired minimal M1 agonism. Relative to prior M1 PAMs that had poor CNS penetration (Kps < 0.1) and cholinergic side effects, we opted for an M1 PAM profile of modest potency, minimal agonism and high CNS penetration. VU319 was efficacious in multiple preclinical cognition models (MEDs of 1 mg/kg PO) and demonstrated no cholinergic AEs in mice (the most sensitive species) or rats, suggesting our strategy was viable. VU319 successfully navigated IND-enabling studies with no alerting genotoxicity, safety pharmacology or repeat-dose oral toxicity liabilities identified at the achieved nonclinical VU-319 doses/exposures, and was devoid of cholinergic AEs in rats, dogs and NHPs. To assess tolerability and pharmacokinetics in humans, VU319 entered a Phase I SAD trial in 2018 (NCT03220295), and was found to be well tolerated with no evidence of cholinergic toxicity (SLUDGE) at doses/exposures where signs of human pro-cognitive benefit were demonstrated. The discovery of the VU319 marks a very important achievement, demonstrating the potential for selective M1 PAMs to engage central M1 receptors in humans without causing cholinergic side effects.
Acknowledgments
The authors thank William K. Warren, Jr. and the William K. Warren Foundation for support of our programs and endowing both the Warren Center for Neuroscience Drug Discovery and the William K. Warren, Jr. Chair in Medicine (C.W.L.).
Glossary
Abbreviations
- PAM
positive allosteric modulator
- PBL
plasma/brain level
- DMPK
drug metabolism and pharmacokinetics
- AE
adverse event
- M1
muscarinic acetylcholine receptor subtype 1
- MED
minimum effective dose
- NOR
novel object recognition
- IND
investigational new drug
- FDA
federal drug administration
Supporting Information Available
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acschemneuro.4c00769.
Additional experimental details, methods for the synthesis and characterization of all compounds, in vitro and in vivo DMPK protocols, and supplemental figures (PDF)
Author Contributions
C.W.L., M.R.W., P.J.C., C.M.N., J.K.R., A.L.B., C.R.H., O.B., A.L.R., and H.P.C. oversaw the medicinal chemistry, target selection and interpreted biological/DMPK data. C.W.L. wrote the manuscript. M.S.P., M.R.W., C.H., S.R.S., J.D.P., B.J.M., and J.L.E. performed chemical synthesis. H.P.C., M.J.N., A.L.R., and C.M.N. performed and analyzed in vitro pharmacology assays. J.W.D., W.P., C.K.J., and J.M.R. performed in vivo behavior pharmacology assays and in vivo DMPK. A.L.B., J.S.D., R.M., R.DC., and T.M.B. performed in vitro and in vivo DMPK studies. M.J.K., A.C., and C.K.J. oversaw IND-enabling studies and development. All authors have given approval to the final version of the manuscript.
Studies were supported by NIH (NIMH, MH082867, MH073676, and MH108498) and the William K. Warren Foundation.
The authors declare the following competing financial interest(s): We are actively developing M1 PAMs with Acadia Pharmaceuticals, although different chemotypes than that disclosed here.
Supplementary Material
References
- Greenlee W.; Clader J.; Asberom T.; McCombie S.; Ford J.; Guzik H.; Kozlowski J.; Li S.; Liu C.; Lowe D.; Vice S.; Zhao H.; Zhou G.; Billard W.; Binch H.; Crosby R.; Duffy R.; Lachowicz J.; Coffin V.; Watkins R.; Ruperto V.; Strader C.; Taylor L.; Cox K. Muscarinic agonists in the treatment of Alzheimer’s disease. Il Farmaco 2001, 56, 247–250. 10.1016/S0014-827X(01)01102-8. [DOI] [PubMed] [Google Scholar]
- Fisher A. Muscarinic receptor agonists in Alzheimer’s disease. CNS Drugs 1999, 12, 197–214. 10.2165/00023210-199912030-00004. [DOI] [Google Scholar]
- Clader J. W. Recent advances in cholinergic drugs for Alzheimer’s disease. Curr. Opin. Drug Discovery Dev. 1999, 2, 311–320. [PubMed] [Google Scholar]
- Brannan S. K.; Sawchak S.; Miller A. C.; Lieberman J. A.; Paul S. M.; Brier A. Muscarinic Cholinergic Receptor Agonist and Peripheral Antagonist for Schizophrenia. N. Engl. J. Med. 2021, 384 (8), 717–726. 10.1056/NEJMoa2017015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melancon B. J.; Tarr J. C.; Panarese J. D.; Wood M. R.; Lindsley C. W. Allosteric modulation of the M1 muscarinic acetylcholine receptor: improving cognition and a potential treatment for schizophrenia and Alzheimer’s disease. Drug Discovery Today 2013, 18 (23–24), 1185–1199. 10.1016/j.drudis.2013.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bridges T. M.; LeBois E. P.; Hopkins C. R.; Wood M. R.; Jones J. K.; Conn P. J.; Lindsley C. W. Antipsychotic potential of muscarinic allosteric modulation. Drug News Perspect. 2010, 23, 229–240. 10.1358/dnp.2010.23.4.1416977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bodick N. C.; Offen W. W.; Levey A. I.; Cutler N. R.; Gauther S. G.; Satlin A.; Shannon H. E.; Tollefson G. D.; Rasmussen K.; Bymaster F. P.; Hurley D. J.; et al. Effects of xanomeline, a selective muscarinic receptor agonist, on cognitive function and behavioral symptoms in Alzheimer disease. Arch. Neurol. 1997, 54 (4), 465–473. 10.1001/archneur.1997.00550160091022. [DOI] [PubMed] [Google Scholar]
- Shekhar A.; Potter W. Z.; Lightfoot J.; Lienemann D.; Dube S.; Mallinckrodt C.; Bymaster F. P.; McKinzie D. L.; Felder C. C. Selective muscarinic receptor agonist xanomeline as a novel treatment approach for schizophrenia. Am. J. Psychiatry 2018, 165, 1033–1039. 10.1176/appi.ajp.2008.06091591. [DOI] [PubMed] [Google Scholar]
- Kaul I.; Sawchak S.; Correll C. U.; Kakar R.; Breier A.; Zhu H.; Miller A. C.; Paul S. M.; Brannan S. K. Efficacy and safety of the muscarinic receptor agonist KarXT (xanomeline–trospium) in schizophrenia (EMERGENT-2) in the USA: results from a randomised, double-blind, placebo-controlled, flexible-dose phase 3 trial. Lancet 2024, 403, 160–170. 10.1016/S0140-6736(23)02190-6. [DOI] [PubMed] [Google Scholar]
- Nguyen H. T. M.; van der Westhuizen E. T.; Langmead C. J.; Tobin A. B.; Sexton P. M.; Christopoulos A.; Valant C. Opportunities and challenges for the development of M1 muscarinic receptor positive allosteric modulators in the treatment for neurocognitive deficits. Br. J. Pharmacol. 2024, 181, 2114–2142. 10.1111/bph.15982. [DOI] [PubMed] [Google Scholar]
- Conn P. J.; Christopolous A.; Lindsley C. W. Allosteric Modulators of GPCRs as a Novel Approach to Treatment of CNS Disorders. Nat. Rev. Drug Discovery 2009, 8, 41–54. 10.1038/nrd2760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lindsley C. W. 2013 Philip S. Portoghese Medicinal Chemistry Lectureship: Drug Discovery Targeting Allosteric Sites. J. Med. Chem. 2014, 57, 7485–7498. 10.1021/jm5011786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shipe W. D.; Lindsley C. W.; Hallett D.. Quinolone M1 receptor positive allosteric modulators. US 8,389,545, 2013.
- Ma L.; Seager M.; Wittman M.; Bickel N.; Burno M.; Jones K.; Graufelds V. K.; Xu G.; Pearson M.; McCampbell A.; Gaspar R.; Shughrue P.; Danzinger A.; Regan C.; Garson S.; Doran S.; Kreatsoulas C.; Veng L.; Lindsley C. W.; Shipe W.; Kuduk S.; Jacobson M.; Sur C.; Kinney G.; Seabrook G. R.; Ray W. J. Selective activation of the M1 muscarinic acetylcholine receptor achieved by allosteric potentiation. Proc. Natl. Acad. Sci. U. S. A. 2009, 106, 15950–15955. 10.1073/pnas.0900903106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shirey J. K.; Brady A. E.; Jones P. J.; Davis A. A.; Bridges T. M.; Jadhav S. B.; Menon U.; Christain E. P.; Doherty J. J.; Quirk M. C.; Snyder D. H.; Levey A. I.; Watson M. L.; Nicolle M. M.; Lindsley C. W.; Conn P. J. A selective allosteric potentiator of the M1 muscarinic acetylcholine receptor increases activity of medial prefrontal cortical neurons and can restore impairments of reversal learning. J. Neurosci. 2009, 29, 14271–14286. 10.1523/JNEUROSCI.3930-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beshore D. C.; Di Marco C. N.; Chang R. K.; Greshock T. J.; Ma L.; Wittman M.; Seager M. A.; Koeplinger K. A.; Thompson C. D.; Fuerst J.; Hartman G. D.; Bilodeau M. T.; Ray W. J.; Kuduk S. D. MK-7622: A first-in-class M1 positive allosteric modulator development candidate. ACS Med. Chem. Lett. 2018, 9, 652–656. 10.1021/acsmedchemlett.8b00095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moran S. P.; Dickerson J. W.; Plumley H. C.; Xiang Z.; Maksymetz J.; Remke D. H.; Doyle C. A.; Niswender C. M.; Engers D. W.; Lindsley C. W.; Rook J. M.; Conn P. J. M1 positive lacking agonist activity provide the optimal profile for enhancing cognition. Neuropsychopharmacology 2018, 43, 1763–1771. 10.1038/s41386-018-0033-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davoren J. E.; Garnsey M.; Pettersen B.; Brodeney M. A.; Edgerton J. R.; Fortin J.-P.; Grimwood S.; Harris A. R.; Jenkison S.; Kenakin T.; Lazzaro J. T.; Lee C.-W.; Lotarski S. M.; Nottebaum L.; O’Neil S. V.; Popiolek M.; Ramsey S.; Steyn S. J.; Thorn C. A.; Zhang L.; Webb D. Design and synthesis of γ- and δ-lactam M1 positive allosteric modulators (PAMs): convulsion and cholinergic toxicity of M1-selective PAM with weak agonist activity. J. Med. Chem. 2017, 60, 6649–6663. 10.1021/acs.jmedchem.7b00597. [DOI] [PubMed] [Google Scholar]
- Moran S. P.; Cho H. P.; Maksymetz J.; Remke D.; Hanson R.; Niswender C. M.; Lindsley C. W.; Rook J. M.; Conn P. J. PF-06827443 displays robust allosteric agonist and positive allosteric modulator activity in high receptor reserve and native systems. ACS Chem. Neurosci. 2018, 9, 2218–2224. 10.1021/acschemneuro.8b00106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rook J. M.; Abe M.; Cho H. P.; Nance K. D.; Luscombe V. B.; Adams J. J.; Dickerson J. W.; Remke D. H.; Garcia-Barrantes P. M.; Engers D. W.; Engers J. L.; Chang S.; Foster J. J.; Blobaum A. L.; Niswender C. M.; Jones C. K.; Conn P. J.; Lindsley C. W. Diverse Effects on M1 Signaling and Adverse Effect Liability within a Series of M1 Ago-PAMs. ACS Chem. Neurosci. 2017, 8, 866–883. 10.1021/acschemneuro.6b00429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rook J. M.; Berton J. L.; Cho H. P.; Gracia-Barrantes P. M.; Moran S. P.; Maksymetz J. T.; Nance K. D.; Dickerosn J. W.; Remke D. H.; Chang S.; Harp J. M.; Blobaum A. L.; Niswender C. M.; Jones C. K.; Stauffer S. R.; Conn P. J.; Lindsley C. W. A novel M1 PAM VU0486846 exerts efficacy in cognition models without displaying agonist activity or cholinergic toxicity. ACS Chem. Neurosci. 2018, 9, 2274–2285. 10.1021/acschemneuro.8b00131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Engers J. L.; Childress E. S.; Long M. F.; Capstick R. A.; Luscombe V. B.; Cho H. P.; Dickerson J. W.; Rook J. M.; Blobaum A. L.; Niswender C. M.; Conn P. J.; Lindsley C. W. VU6007477, a novel M1 PAM based on a pyrrolo[2,3-b]pyridine carboxamide core devoid of cholinergic side effects. ACS Med. Chem. Lett. 2018, 9, 2641–2646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sako Y.; Kurimoto E.; Mandai T.; Suzuki A.; Tanaka M.; Suzuki M.; Shimizu Y.; Yamada M.; Kiruma H. TAK-071, a novel M1 positive allosteric modulator with low cooperativity, improves cognitive function in rodents with few cholinergic side effects. Neuropsychopharmacology 2019, 44, 950–960. 10.1038/s41386-018-0168-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin W.; Mamashli F.; Buhl D. L.; Khudyakov P.; Volfson D.; Martenyi F.; Gevorkyan H.; Rosen L.; Simen A. A. Safety, pharmacokinetics and quantitative EEG modulation of TAK-071, a novel muscarinic M1 receptor positive allosteric modulator, in healthy subjects. Br. J. Pharmacol. 2022, 88, 600–612. 10.1111/bcp.14975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dwomoh L.; Rossi M.; Scarpa M.; Khajehali E.; Molloy C.; Herzyk P.; Bottrill A. R.; Sexton P. M.; Christopoulos A.; Conn P. J.; Lindsley C. W.; Bradley S. J.; Tobin A. B. M1 muscarinic receptor activation reduces pathology and slows the progression of prion-mediated neurodegenerative diseases. Sci. Signal. 2022, 15, eabm3720 10.1126/scisignal.abm3720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Engers J. L.; Smith K. A.; Capstick R. A.; Bender A. M.; Dickerson J. W.; Peng W.; Cho H. P.; Niswender C. M.; Xiang Z.; Blobaum A. L.; Boutaud O.; Rook J. M.; Engers D. W.; Conn P. J.; Lindsley C. W. Discovery of VU6007496: Challenges in the development of an M1 positive allosteric modulator back-up candidate. ACS Chem. Neurosci. 2024, 15, 3421–3433. 10.1021/acschemneuro.4c00508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conley A. C.; Key A. P.; Blackford J. U.; Rook J. M.; Conn P. J.; Lindsley C. W.; Jones C. K.; Newhouse P. A. Cognitive performance effects following a single dose of the M1 muscarinic positive allosteric modulator VU319. Alzheimer’s Dementia 2020, 16, e045339 10.1002/alz.045339. [DOI] [Google Scholar]
- Conley A. C.; Key A. P.; Blackford J. U.; Rook J. M.; Conn P. J.; Lindsley C. W.; Jones C. K.; Newhouse P. A. Functional activity of the muscarinic positive allosteric modulator VU319 during a Phase 1 single ascending dose study. Am. J. Geriatr. Psychiatry 2021, 29, S43. 10.1016/j.jagp.2021.01.038. [DOI] [Google Scholar]
- https://clinicaltrials.gov/study/NCT03220295?term=NCT03220295&rank=1 (accessed Sept 30, 2024).
- Full manuscript detailing the Phase I SAD trial is in preparation.
- Marlo J. E.; Niswender C. M.; Luo Q.; Brady A. E.; Shirey J. K.; Rodriguez A. L.; Bridges T. M.; Williams R.; Days E.; Nalywajko N. T.; Austin C.; Williams M.; Xiang Y.; Orton D.; Brown H. A.; Kim K.; Lindsley C. W.; Weaver C. D.; Conn P. J. Identification and characterization of novel allosteric potentiators of M1 muscarinic receptors reveals multiple modes of activity. Mol. Pharm. 2009, 75, 577–588. 10.1124/mol.108.052886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bridges T. M.; Kennedy J. P.; Cho H. P.; Conn P. J.; Lindsley C. W. Chemical optimization of an M1, M3, M5 positive allosteric modulator (PAM) lead. Part II. Development of a highly selective M1 PAM. Bioorg. Med. Chem. Lett. 2010, 20, 1972–1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melancon B. J.; Poslunsey M. S.; Gentry P. R.; Tarr J. C.; Sheffler D. J.; Mattmann M. E.; Bridges T. M.; Utley T. J.; Daniels J. S.; Niswender C. M.; Conn P. J.; Lindsley C. W.; Wood M. R. Isatin replacements applied to the highly selective, muscarinic M1 PAM ML137: the continued optimization of an MLPCN probe molecule. Bioorg. Med. Chem. Lett. 2013, 23, 412–416. 10.1016/j.bmcl.2012.11.092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghoshal A.; Rook J.; Dickerson J.; Roop G.; Morrison R.; Jalan-Sakrikar N.; Lamsal A.; Noetzel M.; Poslunsey M.; Stauffer S. R.; Xiang Z.; Daniels J. S.; Niswender C. M.; Jones C. K.; Lindsley C. W.; Conn P. J. Selective potentiation of M1 muscarinic receptors reverses deficits in plasticity and negative and cognitive symptoms in repeated phencyclidine treated mouse model of schizophrenia. Neuropsychopharmacology 2016, 41, 598–610. 10.1038/npp.2015.189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- See Supporting Information for full details.
- Lebois E. P.; Bridges T. M.; Dawson E. S.; Kennedy J. p.; Xiang Z.; Jadhav S. B.; Yin H.; Meiler J.; Jones C. K.; Conn P. J.; Weaver C. D.; Lindsley C. W. Discovery and development of novel subtype-selective M1 allosteric agonists for the investigation of M1 receptor function. ACS Chem. Neurosci. 2010, 1, 104–121. 10.1021/cn900003h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Digby G. J.; Noetzel M. J.; Bubser M.; Utley T. J.; Walker A. G.; Byun N. B.; LeBois E. P.; Xiang Z.; Sheffler D. J.; Niswender C. M.; Plumley H. C.; Davis A. A.; Nemirovsky N. E.; Mennenga S.; Camp B. W.; Bimonte-Nelson H. A.; Morrison R.; Daniels S.; Lindsley C. W.; Olive M. F.; Conn P. J. Novel allosteric agonists of the M1 muscarinic acetylcholine receptor induce brain region-specific responses and correspond with behavioral effects in animal models. J. Neurosci. 2012, 32, 8532–8544. 10.1523/JNEUROSCI.0337-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spalding T. A.; Trotter C.; Skjaerbaek N.; Messier T. L.; Currier E. A.; Burstein E. S.; Li D.; Hacksell U.; Brann M. R. Discovery of an ectopic activation site on the M1 muscarinic receptor. Mol. Pharmacol. 2002, 61, 1297–1302. 10.1124/mol.61.6.1297. [DOI] [PubMed] [Google Scholar]
- Hamilton S. E.; Loose M. D.; Qi M.; Levey A. I.; Hille B.; McKnight G. S.; Idzerda R. L.; Nathanson N. M. Disruption of the M1 receptor gene ablates muscarinic receptor-dependent M current regulation and seizure activity in mice. Proc. Natl. Acad. Sci. U. S. A. 1997, 94, 13311–13316. 10.1073/pnas.94.24.13311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Löscher W.; Ferland J.; Ferraro T. N. The relevance of inter- and intrastrain differences in mice and rats and their implications for models of seizures and epilepsy. Epilepsy Behav. 2017, 73, 214–235. 10.1016/j.yebeh.2017.05.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith M.; Arthur B.; Cikowski J.; Holt C.; Gonzalez S.; Fisher N. M.; Vermudez S. A.; Lindsley C. W.; Conn P. J.; Niswender C. M.; Gogliotti R. G. Clinical and preclinical evidence for M1 muscarinic acetylcholine receptor potentiation as a novel therapeutic approach for Rett syndrome. Neurotherapeutics 2022, 19, 1340–1352. 10.1007/s13311-022-01254-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vecchio I.; Sorrentino L.; Paoletti A.; Marra R.; Arbitrio M. The state of the art of acetylcholinesterase inhibitors in the treatment of Alzheimer’s disease. J. Cent. Nerv. Syst. Dis. 2021, 13, 11795735211029113 10.1177/11795735211029113. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.










