Abstract

Since the 1950s, understanding of antipsychotic activity in schizophrenia has been largely grounded in the dopamine (DA) hypothesis. Most antipsychotics approved for schizophrenia interact with D2 DA receptors as an important part of their mechanism of action. While antipsychotics blocking D2 DA receptors can be effective for positive symptoms of schizophrenia, none are approved by regulatory authorities for predominant negative or cognitive symptoms. Moreover, many of these agents induce a range of problematic side effects related to D2 DA receptor blockade (eg, drug-induced parkinsonism, akathisia, tardive dyskinesia, hyperprolactinemia and related sexual side effects, sedation). This has prompted the search for novel mechanisms with improved efficacy and tolerability based on evidence supporting involvement of other neurotransmitter systems in schizophrenia pathophysiology, including acetylcholine, gamma-aminobutyric acid, and glutamate. Among these options, targeting muscarinic receptors emerged as a promising treatment strategy. In September 2024, the U.S. Food and Drug Administration approved xanomeline and trospium chloride for treatment of adults with schizophrenia based on results from three 5-week, randomized, double-blind, placebo-controlled trials and two 52-week open-label trials. In the placebo-controlled trials, xanomeline/trospium reduced symptoms of schizophrenia, was generally well tolerated, and was not associated with clinically meaningful motor symptoms, hyperprolactinemia, sexual side effects, or weight gain compared with placebo. The long-term safety of xanomeline/trospium was also confirmed in two 52-week, open-label trials. This paper reviews the preclinical and clinical rationale for muscarinic receptor activation as a treatment for schizophrenia and the efficacy, safety, and tolerability profile of xanomeline/trospium.

INTRODUCTION

Schizophrenia is a serious and heterogeneous mental health disorder affecting approximately 24 million people worldwide and associated with substantial disability, morbidity, and mortality.1,2 The positive (eg, delusions, hallucinations, disorganized speech and behavior), negative (eg, diminished expression, alogia, anhedonia, avolition), and cognitive (eg, impaired attention, working memory, processing speed, executive function) symptoms of schizophrenia contribute to functional and psychosocial impairment. People living with schizophrenia experience substantially more disability than those with anxiety disorders, major depressive disorder, bipolar disorder, alcohol use disorder, or autism spectrum disorder,3 making it one of the top 15 causes of disability worldwide.4 Life expectancy of people living with schizophrenia is also reduced by almost 30 years compared with the general population.5 Among the contributors to the excess mortality of schizophrenia are unnatural causes (eg, suicide, accidents) and medical comorbidities (eg, cardiovascular disease and diabetes mellitus).5-9

Since the 1950s, understanding of schizophrenia pathogenesis and antipsychotic activity has been largely grounded in various iterations of the dopamine (DA) hypothesis. The core aspects of the DA hypothesis posit that DA dysregulation in various brain regions, including the limbic and associative striatum and prefrontal cortex (PFC), contributes in part to the symptoms of schizophrenia.10,11 The hypothesis that excessive presynaptic DA release into relevant areas of the striatum underlies the positive symptoms of schizophrenia explains the efficacy basis of most antipsychotics approved prior to 2024: positive symptoms were lessened by postsynaptic D2 DA receptor antagonism or partial antagonism.12 Unlike most antipsychotics, the efficacy of clozapine is likely to involve other receptors beyond D2 DA receptors. Although clozapine has lower binding affinity for D2 DA receptors, it can be effective for patients with inadequate response to D2 DA receptor binding antipsychotics (ie, those with treatment-resistant schizophrenia) and this appears to be due to non-dopaminergic activities that contribute to its clinical profile.13

Current antipsychotics are helpful for many people living with schizophrenia but have numerous limitations. Up to 80% of people have residual positive symptoms despite adequate doses and duration of antipsychotic treatment14; moreover, no antipsychotics are approved for the treatment of predominant negative or cognitive symptoms, and D2 DA receptor or anticholinergic activities can worsen negative and cognitive symptoms (eg, attention, memory, executive function, processing speed).15,16 Antipsychotic side effects directly related to D2 DA receptor antagonism include motor symptoms (drug-induced parkinsonism [DIP], akathisia, tardive dyskinesia [TD]) and hyperprolactinemia and related sexual AEs, all of which contribute to poor adherence and relapse risk.17,18 Atypical antipsychotics are also associated with metabolic AEs including dyslipidemia, glucose intolerance, and weight gain.19 Early use of reserpine and tetrabenazine in the 1950s and 1960s, both of which deplete catecholamines from presynaptic vesicles by vesicular monoamine transporter type 2 (VMAT2) inhibition20 generated long-standing interest in modulating presynaptic DA release in schizophrenia rather than managing its sequelae by blocking postsynaptic D2 DA receptors. Unfortunately, VMAT2-induced presynaptic DA depletion was nonselective, impacting striatal regions associated with positive symptoms and motor areas alike and resulting in substantial rates of DIP and akathisia at doses that controlled psychotic symptoms.21,22

The search for presynaptic mechanisms was not entirely abandoned, especially when advances in neuroimaging techniques supported that positive symptoms were indeed a presynaptic problem in most individuals with schizophrenia, and arose from excess release of DA in the human associative striatum (and adjacent portions of the sensorimotor striatum). These findings parallel the presynaptic DA dysfunction found in the mesolimbic pathway in animal models (Figure 1).23 While DA dysfunction remains central to the pathophysiology of schizophrenia, evidence supporting involvement of additional neurotransmitter systems, including acetylcholine (ACh),25-28 gamma-aminobutyric acid (GABA),29,30 serotonin,31 and glutamate,31-34 provided the stimulus to explore a range of strategies that might address schizophrenia symptoms without D2 DA receptor binding.23 Presynaptic DA depletion was largely abandoned in the early 1960s due to the significant complaints of motor adverse effects and depression seen with reserpine and tetrabenazine35; however, another approach to modulating presynaptic DA release that was also selective for striatal pathways related to positive symptoms was discovered in the form of muscarinic receptor agonism. The potential for this approach is highlighted by the recent approval of xanomeline and trospium chloride by the U.S. Food and Drug Administration (FDA) in September 2024. Here, we review the rationale for targeting muscarinic receptors in schizophrenia and key evidence from xanomeline/trospium clinical trials that supports the validity and feasibility of this approach (see Paul26 and Dean36 for review of other approaches for targeting muscarinic receptors).

Recent advances in understanding of striatal connectivity.23 (A) Summary of primate tracing studies mapping connections between the cortex, striatum, and midbrain. Tract tracing studies showed that striatocortical connections run in 3 parallel pathways: motor areas project to the caudal putamen, dorsolateral prefrontal cortex to caudate and rostral putamen, and limbic areas to the ventral striatum. These subdivisions are termed the sensorimotor, associative, and limbic striatum. Subsequent research using retrograde tracers injected into striatum to determine midbrain connections showed that VTA projects primarily to limbic striatum, while dorsomedial/ventrolateral parts of the SN project to the associative and sensorimotor striatum, respectively. In primates, the VTA is proportionally smaller than in rodents; it innervates the ventral striatum, whereas the dorsomedial SN innervates the associative striatum, and the ventrolateral SN innervates the sensorimotor striatum. The striatum in turn has efferents projecting back to the midbrain. In addition to these reciprocal connections, feedforward striatonigrostriatal connections allow information to pass along the striatum from limbic to motor regions via the associative striatum. Human imaging studies confirm that the positive symptoms of psychosis in patients with schizophrenia are associated with increased DA activity in the associative and adjacent portions of the sensorimotor striatum that receive input from the dorsomedial SN. Importantly, dopaminergic overactivity was not seen in limbic striatum. (B) Summary of rodent–primate differences in mesostriatal connectivity. In rodents, the ventral striatum is proportionally larger than in primates. The NAcc shell is innervated by the medial VTA, the NAcc core by the central VTA, and the lateral VTA innervates a region homologous to the associative striatum. Together these connections from VTA define the mesolimbic or A10 pathway. The medial SN has connections to more dorsal aspects of the ventral striatum, while lateral SN projects to the dorsal motor regions of the striatum. (C) Modern neurochemical imaging findings suggest that it is within dorsal regions of the striatum that dopaminergic aberrations are greatest.24 *P < .05. NAcc, nucleus accumbens; om, orbital/medial; PFC, prefrontal cortex; SN, substantia nigra; vm, ventromedial; VTA, ventral tegmental area.
Figure 1.

Recent advances in understanding of striatal connectivity.23 (A) Summary of primate tracing studies mapping connections between the cortex, striatum, and midbrain. Tract tracing studies showed that striatocortical connections run in 3 parallel pathways: motor areas project to the caudal putamen, dorsolateral prefrontal cortex to caudate and rostral putamen, and limbic areas to the ventral striatum. These subdivisions are termed the sensorimotor, associative, and limbic striatum. Subsequent research using retrograde tracers injected into striatum to determine midbrain connections showed that VTA projects primarily to limbic striatum, while dorsomedial/ventrolateral parts of the SN project to the associative and sensorimotor striatum, respectively. In primates, the VTA is proportionally smaller than in rodents; it innervates the ventral striatum, whereas the dorsomedial SN innervates the associative striatum, and the ventrolateral SN innervates the sensorimotor striatum. The striatum in turn has efferents projecting back to the midbrain. In addition to these reciprocal connections, feedforward striatonigrostriatal connections allow information to pass along the striatum from limbic to motor regions via the associative striatum. Human imaging studies confirm that the positive symptoms of psychosis in patients with schizophrenia are associated with increased DA activity in the associative and adjacent portions of the sensorimotor striatum that receive input from the dorsomedial SN. Importantly, dopaminergic overactivity was not seen in limbic striatum. (B) Summary of rodent–primate differences in mesostriatal connectivity. In rodents, the ventral striatum is proportionally larger than in primates. The NAcc shell is innervated by the medial VTA, the NAcc core by the central VTA, and the lateral VTA innervates a region homologous to the associative striatum. Together these connections from VTA define the mesolimbic or A10 pathway. The medial SN has connections to more dorsal aspects of the ventral striatum, while lateral SN projects to the dorsal motor regions of the striatum. (C) Modern neurochemical imaging findings suggest that it is within dorsal regions of the striatum that dopaminergic aberrations are greatest.24 *P < .05. NAcc, nucleus accumbens; om, orbital/medial; PFC, prefrontal cortex; SN, substantia nigra; vm, ventromedial; VTA, ventral tegmental area.

RATIONALE FOR TARGETING MUSCARINIC RECEPTORS IN SCHIZOPHRENIA

The unexpected realization in the late 1990s that muscarinic receptor activation was another method for regulating presynaptic DA release revived a hypothesized connection that remained unexplored and incompletely understood for decades.26,27 In 1957, data were published indicating that the nonselective pan muscarinic receptor agonist arecoline possessed antipsychotic activity in preclinical models of psychosis and in clinical studies.37 Unfortunately, these early arecoline studies in schizophrenia were largely forgotten or ignored in the wake of chlorpromazine’s discovery and the subsequent hyperfocus on the DA hypothesis of psychosis.28 In the 1980s, muscarinic receptor agonists, including xanomeline, a potent synthetic muscarinic receptor agonist derived from arecoline, were developed as potential procognitive drugs in neurocognitive disorders such as dementia.25,27 At the time of its synthesis, xanomeline was initially characterized as an M1 muscarinic receptor agonist.38 Subsequent work demonstrated that xanomeline binds to and has functional activity at all 5 muscarinic receptor subtypes but with the highest degree of intrinsic activity at M1 and M4 muscarinic receptors.39,40

The revived interest in muscarinic receptor agonists as a potential antipsychotic strategy stems from a serendipitous finding seen in a clinical trial of xanomeline for people with Alzheimer’s disease (AD). In a 6-month, double-blind, placebo-controlled phase 2 clinical trial enrolling 343 participants, xanomeline improved cognition (measured by the cognitive subscale of the Alzheimer’s Disease Assessment Scale) and was also associated with dose-dependent and rapid improvements in psychotic symptoms (measured by the Alzheimer’s Disease Symptomatology Scale) in people with these symptoms at trial baseline, and also mitigated new onset positive symptoms.41 In 2008, xanomeline was subsequently evaluated in a 4-week, double-blind, placebo-controlled schizophrenia trial (N = 20). Despite the small sample size and limited statistical power, xanomeline was associated with significant improvements in psychotic symptoms (measured by the Positive and Negative Syndrome Scale [PANSS] and the Brief Psychiatric Rating Scale) and also demonstrated procognitive effects in several subtests of the neuropsychological test battery.42 However, in both the dementia and schizophrenia trials, xanomeline was associated with substantial peripheral procholinergic side effects (eg, nausea, vomiting) that stymied further clinical development.41-43

Prior to the 2008 schizophrenia trial, xanomeline’s pharmacology was explored extensively in preclinical models to further understand the basis of its antipsychotic activity.44-47 In vitro studies noted that xanomeline did not bind to D2 DA receptors, so antipsychotic activity was necessarily unrelated to any direct DA receptor interaction. In vivo work noted that xanomeline inhibited amphetamine-induced activity in a mouse model of psychosis; however, xanomeline’s activity was substantially attenuated in M4 muscarinic receptor knockout mice and was attenuated in M1 muscarinic receptor knockout mice, demonstrating that its effects at reducing hyperdopaminergic behaviors required activation of these receptor subtypes.48 The antipsychotic effects of xanomeline were also blocked by central muscarinic receptor antagonists such as scopolamine.44 Conversely, xanomeline reversed scopolamine-induced disruptions in latent inhibition in a rat model of schizophrenia.49 Taken together, these findings support that the antipsychotic effects of xanomeline are mediated by stimulation of muscarinic receptors.

MUSCARINIC RECEPTOR PHARMACOLOGY

Spurred by the fortuitous discovery of xanomeline’s antipsychotic activity, research accelerated greatly over the next 30 years focusing on muscarinic receptor biology and its potential connection to schizophrenia symptoms. Muscarinic receptors belong to the superfamily of G protein–coupled receptors and include 5 distinct subtypes (M1-M5) grouped into 2 classes based on their cognate G proteins and location within the synapse. The M1, M3, and M5 subtypes primarily mediate excitatory postsynaptic currents via coupling to Gq proteins, while the M2 and M4 subtypes are predominantly presynaptic and inhibit depolarization through activation of Gi/o proteins.27 Muscarinic receptors play a role in neuronal excitability and neurotransmitter regulation, learning and memory, cardiac function, smooth muscle contraction, and exocrine gland secretion (eg, saliva, gastric acid)27 and are widely expressed throughout the central and peripheral nervous system.27,50

Of particular relevance to this review, both M1 and M4 muscarinic receptors are highly expressed in brain regions implicated in cognitive function and the positive symptoms of psychosis, particularly those regions containing key DA circuitry (eg, PFC, limbic striatum, associative striatum).28,51 In rodents, expression of D2 DA and M1/M4 muscarinic receptors overlaps in the nucleus accumbens, the terminal end of the mesolimbic tract that originates in the ventral tegmental area (VTA). Overactivation of these DA afferents promotes excessive presynaptic DA release and subsequent postsynaptic D2 DA receptor activation associated with positive psychotic symptoms.27 M1 muscarinic receptors are expressed at high levels in the cortex and hippocampus, but not so for M4 muscarinic receptors or D2 DA receptors.27,52 Data suggest that M4 muscarinic receptor expression is decreased in the hippocampus among people with schizophrenia.53,54 Notably, M1 and M4 muscarinic receptors are not highly expressed in brain regions related to D2 DA blockade-mediated AEs (eg, tuberoinfundibular system, sensorimotor striatum).27 See Scarr55,56 and Perry57 for additional information on the cholinergic system in the human central nervous system (CNS).

Preclinical research confirmed that knockout mice lacking either the M1 or M4 muscarinic receptor exhibit psychosis-like phenotypes.58-60 In wild-type mice, increasing ACh signaling with M1 or M4 muscarinic receptor orthosteric agonists or positive allosteric modulators (PAMs) blocked the psychotomimetic effects of N-methyl D-aspartate (NMDA) antagonists (eg, phenylcyclohexyl piperidine [PCP], MK-801).28,50 Consistent with these findings, polymorphisms in both M1 and M4 muscarinic receptors have been found in people with schizophrenia and are associated with clinical manifestations including dysregulated error processing and reduced gray matter volume.61-63 Studies have also noted that individuals with schizophrenia have lower M1 muscarinic receptor levels and expression than peers without schizophrenia.64,65 Moreover, approximately 25% of those with schizophrenia have 75% lower M1 muscarinic receptor expression than expected, a finding described as the muscarinic receptor deficit syndrome (MRDS)66,67 (Figure 2). Low M1 muscarinic receptor expression by itself may not always correlate with cognitive impairment but this remains a potential target to improve cognition in schizophrenia.

The probability distribution of cortical M1muscarinic receptor levels in people living with schizophrenia and controls.66 The probability distribution of cortical M1 muscarinic receptor levels measured using [3H]pirenzepine binding in 80 people with schizophrenia (blue line) and 74 controls (red line). The kernel density estimation of these binding data showed that people with schizophrenia consisted of 2 groups and the controls 1 group. One group in the schizophrenia group consisted of 26 people with a marked deficit in M1 muscarinic receptor (muscarinic receptor deficit schizophrenia: MRDS). In the second group, levels of cortical M1 muscarinic receptors were numerically lower but not statistically significantly different from controls (non-MRDS). ©2020 Dean B, Scarr E. Psychiatry Res. 2020; 288:112989. Used with permission from Elsevier. 
Figure 2.

The probability distribution of cortical M1muscarinic receptor levels in people living with schizophrenia and controls.66 The probability distribution of cortical M1 muscarinic receptor levels measured using [3H]pirenzepine binding in 80 people with schizophrenia (blue line) and 74 controls (red line). The kernel density estimation of these binding data showed that people with schizophrenia consisted of 2 groups and the controls 1 group. One group in the schizophrenia group consisted of 26 people with a marked deficit in M1 muscarinic receptor (muscarinic receptor deficit schizophrenia: MRDS). In the second group, levels of cortical M1 muscarinic receptors were numerically lower but not statistically significantly different from controls (non-MRDS). ©2020 Dean B, Scarr E. Psychiatry Res. 2020; 288:112989. Used with permission from Elsevier. 

RATIONALE FOR TARGETING MUSCARINIC RECEPTORS FOR POSITIVE SYMPTOMS OF PSYCHOSIS IN SCHIZOPHRENIA

M4 Muscarinic Receptors

As previously noted, animal models of positive symptoms of psychosis note increased activity of VTA DA neurons resulting in heightened terminal DA release in the nucleus accumbens, while in humans, the increased DA activity occurs in an analogous pathway that connects the dorsomedial substantia nigra to the associative striatum (Figure 3A).27,68,74 The clinical finding of xanomeline’s antipsychotic properties propelled research with animal models to identify the role of cholinergic pathways that relate to positive symptoms. This research uncovered the existence of brainstem afferent cholinergic neurons that originate in the laterodorsal tegmentum (LDT) and project to the VTA. Release of ACh from the LDT activates M5 muscarinic receptors on the VTA cell body, thereby stimulating striatal DA release.27,68

(A) Hypothesized role of M4muscarinic receptor activation in rodent models of psychosis.M4muscarinic receptors.27,68-70(B) Hypothesized role of M1muscarinic receptor activation in rodent models of psychosis.27,70-73 Afferent cholinergic neurons project from the LDT to the medial VTA where they synapse onto DA neurons that project to the striatum. These DA neurons also receive input from glutamatergic pyramidal neurons originating in the PFC that are in turn regulated by parvalbumin-positive GABAergic interneurons. M4 autoreceptors are expressed presynaptically on the cholinergic projections in the medial VTA. Activation of M4 autoreceptors reduces cholinergic signaling to the VTA, inhibiting VTA DA release into the striatum and reducing positive psychotic symptoms. M1 muscarinic receptors are expressed on the parvalbumin-positive GABAergic neurons in the PFC. Activation of these excitatory receptors increases GABA-mediated inhibition of glutamatergic signaling from the PFC to the medial VTA. Decreased glutamate release reduces DA neuron activation in the VTA and DA release in the striatum. ACh, acetylcholine; DA, dopamine; GABA, gamma-aminobutyric acid; Glu, glutamate; LDT, laterodorsal tegmentum; PFC, prefrontal cortex; VTA, ventral tegmental area.
Figure 3.

(A) Hypothesized role of M4muscarinic receptor activation in rodent models of psychosis.M4muscarinic receptors.27,68-70(B) Hypothesized role of M1muscarinic receptor activation in rodent models of psychosis.27,70-73 Afferent cholinergic neurons project from the LDT to the medial VTA where they synapse onto DA neurons that project to the striatum. These DA neurons also receive input from glutamatergic pyramidal neurons originating in the PFC that are in turn regulated by parvalbumin-positive GABAergic interneurons. M4 autoreceptors are expressed presynaptically on the cholinergic projections in the medial VTA. Activation of M4 autoreceptors reduces cholinergic signaling to the VTA, inhibiting VTA DA release into the striatum and reducing positive psychotic symptoms. M1 muscarinic receptors are expressed on the parvalbumin-positive GABAergic neurons in the PFC. Activation of these excitatory receptors increases GABA-mediated inhibition of glutamatergic signaling from the PFC to the medial VTA. Decreased glutamate release reduces DA neuron activation in the VTA and DA release in the striatum. ACh, acetylcholine; DA, dopamine; GABA, gamma-aminobutyric acid; Glu, glutamate; LDT, laterodorsal tegmentum; PFC, prefrontal cortex; VTA, ventral tegmental area.

Neurophysiology studies later discovered that ACh release from LDT afferents was regulated by inhibitory M4 autoreceptors on axonal terminals; therefore, activation of these inhibitory autoreceptors by M4 muscarinic receptor agonists or PAMs reduces LDT ACh release, resulting in less cholinergic stimulation of VTA DA neurons.68 Taken together, the net biological effect of M4 muscarinic receptor activation is decreased VTA-mediated release of DA in the striatum and a reduction in positive/psychosis symptoms. The absence of motor effects from M4 muscarinic receptor activation is explained by the fact that in dorsal striatal motor areas, presynaptic DA release is regulated by another cholinergic pathway, the pedunculopontine nucleus (PPN). On cholinergic PPN neurons, activation of muscarinic M2 inhibitory autoreceptors is necessary to reduce cholinergic output.75 The finding of distinct muscarinic autoreceptor control of brainstem cholinergic inputs to striatal DA circuits provides the intriguing option of reducing positive psychosis symptoms by selective actions on relevant DA neurons while sparing off-target effects on motoric DA pathways.

M1 Muscarinic Receptors

The mechanism by which M1 muscarinic receptor activation might modulate neural circuits implicated in the positive symptoms of psychosis is hypothesized to be the product of cortical M1 muscarinic heteroreceptors mediating “top-down” glutamatergic modulation of VTA DA release (Figure 3B).27 Parvalbumin-positive GABAergic interneurons in the PFC regulate the activity of glutamatergic pyramidal neurons, and these inhibitory interneurons express M1 muscarinic receptors.27,71,72 These descending glutamate pathways terminate in the VTA, where glutamate signals stimulate presynaptic VTA striatal DA release. Activation of excitatory M1 muscarinic receptors on GABAergic interneurons facilitates GABA release and, in turn, downregulates glutamate signaling.27 The net effect is diminished glutamate release onto the VTA, decreased VTA activation, and less DA release in the striatum.27 M1 muscarinic receptor activation thus provides another presynaptic mechanism by which positive symptoms of psychosis can be reduced. M1 muscarinic receptors in the human cortex are primarily found on pyramidal cell bodies.76 Compared with controls, diminished cortical levels of M1-expressing neurons are seen in people with schizophrenia. These observations suggest that changes in cortical M1 muscarinic receptor levels among individuals with schizophrenia may affect glutamate homeostasis (see Dean66 for review).

RATIONALE FOR TARGETING MUSCARINIC RECEPTORS FOR COGNITIVE IMPAIRMENT IN SCHIZOPHRENIA

One basis for cognitive impairment in schizophrenia relates to an altered balance of excitatory (glutamate) and inhibitory (GABA) neurotransmission leading to disinhibition and discoordination of interacting neural networks.51,77 Converging lines of evidence from in vitro and in vivo studies suggest that M1 and M4 muscarinic receptors may be important therapeutic targets for the treatment of cognitive impairment in schizophrenia in part due to their interactions with glutamatergic circuits.27,51 Abnormal cholinergic neurotransmission also contributes to cognitive dysfunction based on the findings noted previously: schizophrenia is associated with low M1 receptor expression, with 25% of patients having MRDS (ie, M1 muscarinic receptor expression more than 75% below mean levels in the general population).66,67

M4 Muscarinic Receptors

The appropriate balance between excitatory and inhibitory signaling in the hippocampus is critical to learning and memory processes, and disruption of this balance can lead to cognitive impairment.78,79 M4 muscarinic receptors are expressed on excitatory pyramidal cells in the CA3 area of the hippocampus and may have a role in modulating this balance.80 These pyramidal cells control excitatory drive to CA1 by releasing glutamate at the CA3-CA1 synapse. Activation of M4 muscarinic receptors in CA3 lowers excitatory drive and is required for cholinergic suppression of neurotransmission at this synapse (Figure 4B).80,84

Hypothesized role of muscarinic receptor activation in cognitive impairment in schizophrenia. (A) M1muscarinic receptors: modulating excitatory/inhibitory balance in the prefrontal cortex.70,71,81-83(B) M4muscarinic receptors: modulating excitatory/inhibitory imbalance in the hippocampus.80,84 Cognitive deficits in schizophrenia may arise from an altered balance of excitatory (glutamate) and inhibitory (GABA) neurotransmission in multiple brain regions including the PFC and hippocampus. M1 muscarinic receptors are expressed on GABAergic interneurons in the PFC that synapse onto glutamatergic neurons. Therefore, activation of these receptors increases GABA release and may normalize the excitatory/inhibitory balance in cortical areas associated with cognition. In the hippocampus, M4 muscarinic receptors are expressed on CA3 excitatory (glutamatergic) pyramidal cells that project to the CA1 area. Activation of M4 muscarinic receptors in CA3 lowers excitatory drive and is required for cholinergic suppression of neurotransmission at the CA3-to-CA1 synapse. GABA, gamma-aminobutyric acid; Glu, glutamate; PFC, prefrontal cortex.
Figure 4.

Hypothesized role of muscarinic receptor activation in cognitive impairment in schizophrenia. (A) M1muscarinic receptors: modulating excitatory/inhibitory balance in the prefrontal cortex.70,71,81-83(B) M4muscarinic receptors: modulating excitatory/inhibitory imbalance in the hippocampus.80,84 Cognitive deficits in schizophrenia may arise from an altered balance of excitatory (glutamate) and inhibitory (GABA) neurotransmission in multiple brain regions including the PFC and hippocampus. M1 muscarinic receptors are expressed on GABAergic interneurons in the PFC that synapse onto glutamatergic neurons. Therefore, activation of these receptors increases GABA release and may normalize the excitatory/inhibitory balance in cortical areas associated with cognition. In the hippocampus, M4 muscarinic receptors are expressed on CA3 excitatory (glutamatergic) pyramidal cells that project to the CA1 area. Activation of M4 muscarinic receptors in CA3 lowers excitatory drive and is required for cholinergic suppression of neurotransmission at the CA3-to-CA1 synapse. GABA, gamma-aminobutyric acid; Glu, glutamate; PFC, prefrontal cortex.

Disruptions in this pathway are associated with cognitive deficits in preclinical models. M4 muscarinic receptor knockout mice have deficits in the acquisition of both contextual and cue-dependent fear conditioning but not spatial memory, suggesting that M4 muscarinic receptors may have a role in certain types of memory.60,85 Conversely, M4 muscarinic receptor agonists have shown procognitive benefits in rodent models85-88 and in nonhuman primates.89 In rodent imaging studies, administration of an M4 muscarinic receptor PAM can normalize amphetamine-induced changes in hippocampal activity90 and enhance the rate of acquisition,87 suggesting modulation of M4 muscarinic receptors can enhance cognition.51

M1 Muscarinic Receptors

M1 muscarinic receptors are highly expressed in the cortex and hippocampus27 and are hypothesized to play a central role in learning, memory, and executive functions.51 In addition to direct stimulation of cholinergic neurotransmission, activation of M1 heteroreceptors expressed on PFC GABAergic interneurons increases inhibitory GABA signaling, as described previously.27,71,72 Increased GABA release onto glutamate pathways not only can moderate subcortical processes (eg, glutamate stimulation of VTA DA release), but activation of parvalbumin-positive GABAergic interneurons is also hypothesized to normalize the excitatory/inhibitory balance in cortical areas associated with cognition (Figure 4A).27,77

The proposed models described above are supported by multiple preclinical and human studies. Loss of cholinergic neurons, particularly in the nucleus basalis of Meynert, is implicated in the pathophysiology of cognitive impairment in neurodegenerative disorders such as Parkinson’s Disease and AD.91 In rodent models, pharmacological blockade or genetic deletion of M1 muscarinic receptors results in significant learning and memory disturbances.92-95 Conversely, in vivo studies found that activation of M1 muscarinic receptors enhances synaptic plasticity in prefrontal and hippocampal circuits,80,96 increases neuronal excitability,97 facilitates learning, and improves cognitive deficits induced by NMDA antagonists50 or genetic mutations in the NMDA receptor.98 Furthermore, M1 muscarinic receptor activation has been shown to enhance memory consolidation and retrieval in various tasks (eg, object recognition, spatial learning, fear conditioning), executive function and attention,99 and cognitive flexibility.100

OVERVIEW OF XANOMELINE/TROSPIUM CLINICAL DEVELOPMENT PROGRAM

In September 2024, the U.S. FDA approved xanomeline/trospium, making it the first approved muscarinic receptor agonist for the treatment of schizophrenia. This medication combines the dual M1/M4 muscarinic receptor agonist xanomeline and the peripherally restricted muscarinic receptor antagonist trospium chloride. Xanomeline is responsible for efficacy, while the role of trospium is to reduce side effects associated with peripheral receptor activation by xanomeline without interfering with xanomeline’s mechanism in the CNS. The basis for the choice of trospium rests in the fact that it possesses a highly polar quaternary amine structure that limits passive diffusion into the CNS while also having high affinity for the P-glycoprotein efflux transporter present at the blood-brain barrier, which leads to its rapid extrusion from the CNS.101,102 The strategy of pairing xanomeline with trospium was subsequently evaluated in a double-blind, randomized, placebo-controlled trial; xanomeline paired with trospium demonstrated a clinically meaningful improvement in tolerability compared with xanomeline paired with placebo.103 A global clinical development program was then undertaken to evaluate the potential of xanomeline/trospium in schizophrenia as monotherapy or adjunctive therapy to other antipsychotics, and as monotherapy for psychosis in AD (Table 1).

Table 1.

Xanomeline/trospium core clinical development program.

TrialDevelopment phasePopulationTrial descriptionStatus
EMERGENT-1Phase 2Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT03697252)
Completed, results published
EMERGENT-2Phase 3Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT04659161)
Completed, results published
EMERGENT-3Phase 3Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT04738123)
Completed, results published
EMERGENT-4Phase 3Schizophrenia52-week, open-label extension trial of EMERGENT-2 and EMERGENT-3
(long-term safety; NCT04659174)
Completed
EMERGENT-5Phase 3Schizophrenia52-week, open-label trial
(long-term safety; NCT04820309)
Completed
ARISEPhase 3Schizophrenia6-week, randomized, double-blind, placebo-controlled trial of adjunctive xanomeline/trospium in people with inadequately controlled symptoms of schizophrenia (efficacy and safety of adjunctive xanomeline/trospium; NCT051453413)Ongoing
ARISE OLEPhase 3Schizophrenia52-week, outpatient, open-label extension trial of ARISE
(long-term safety and tolerability of adjunctive xanomeline/trospium; NCT05304767)
Ongoing
ADEPT-1Phase 3Psychosis in Alzheimer’s Disease12-week, single-blind treatment period followed by 26-week, double-blind,
placebo-controlled, randomized withdrawal period
(relapse prevention; NCT05511363)
Ongoing
ADEPT-2Phase 3Psychosis in Alzheimer’s Disease12-week, double-blind, placebo-controlled trial
(acute efficacy; NCT06126224)
Ongoing
ADEPT-3Phase 3Psychosis in Alzheimer’s Disease52-week, open-label extension trial of ADEPT-1 and ADEPT-2
(long-term efficacy and safety; NCT05980949)
Ongoing
ADEPT-4Phase 3Psychosis in Alzheimer’s Disease12-week, double-blind, placebo-controlled trial (acute efficacy; NCT06585787)Ongoing
TrialDevelopment phasePopulationTrial descriptionStatus
EMERGENT-1Phase 2Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT03697252)
Completed, results published
EMERGENT-2Phase 3Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT04659161)
Completed, results published
EMERGENT-3Phase 3Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT04738123)
Completed, results published
EMERGENT-4Phase 3Schizophrenia52-week, open-label extension trial of EMERGENT-2 and EMERGENT-3
(long-term safety; NCT04659174)
Completed
EMERGENT-5Phase 3Schizophrenia52-week, open-label trial
(long-term safety; NCT04820309)
Completed
ARISEPhase 3Schizophrenia6-week, randomized, double-blind, placebo-controlled trial of adjunctive xanomeline/trospium in people with inadequately controlled symptoms of schizophrenia (efficacy and safety of adjunctive xanomeline/trospium; NCT051453413)Ongoing
ARISE OLEPhase 3Schizophrenia52-week, outpatient, open-label extension trial of ARISE
(long-term safety and tolerability of adjunctive xanomeline/trospium; NCT05304767)
Ongoing
ADEPT-1Phase 3Psychosis in Alzheimer’s Disease12-week, single-blind treatment period followed by 26-week, double-blind,
placebo-controlled, randomized withdrawal period
(relapse prevention; NCT05511363)
Ongoing
ADEPT-2Phase 3Psychosis in Alzheimer’s Disease12-week, double-blind, placebo-controlled trial
(acute efficacy; NCT06126224)
Ongoing
ADEPT-3Phase 3Psychosis in Alzheimer’s Disease52-week, open-label extension trial of ADEPT-1 and ADEPT-2
(long-term efficacy and safety; NCT05980949)
Ongoing
ADEPT-4Phase 3Psychosis in Alzheimer’s Disease12-week, double-blind, placebo-controlled trial (acute efficacy; NCT06585787)Ongoing

Abbreviation: OLE, open-label extension.

Table 1.

Xanomeline/trospium core clinical development program.

TrialDevelopment phasePopulationTrial descriptionStatus
EMERGENT-1Phase 2Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT03697252)
Completed, results published
EMERGENT-2Phase 3Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT04659161)
Completed, results published
EMERGENT-3Phase 3Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT04738123)
Completed, results published
EMERGENT-4Phase 3Schizophrenia52-week, open-label extension trial of EMERGENT-2 and EMERGENT-3
(long-term safety; NCT04659174)
Completed
EMERGENT-5Phase 3Schizophrenia52-week, open-label trial
(long-term safety; NCT04820309)
Completed
ARISEPhase 3Schizophrenia6-week, randomized, double-blind, placebo-controlled trial of adjunctive xanomeline/trospium in people with inadequately controlled symptoms of schizophrenia (efficacy and safety of adjunctive xanomeline/trospium; NCT051453413)Ongoing
ARISE OLEPhase 3Schizophrenia52-week, outpatient, open-label extension trial of ARISE
(long-term safety and tolerability of adjunctive xanomeline/trospium; NCT05304767)
Ongoing
ADEPT-1Phase 3Psychosis in Alzheimer’s Disease12-week, single-blind treatment period followed by 26-week, double-blind,
placebo-controlled, randomized withdrawal period
(relapse prevention; NCT05511363)
Ongoing
ADEPT-2Phase 3Psychosis in Alzheimer’s Disease12-week, double-blind, placebo-controlled trial
(acute efficacy; NCT06126224)
Ongoing
ADEPT-3Phase 3Psychosis in Alzheimer’s Disease52-week, open-label extension trial of ADEPT-1 and ADEPT-2
(long-term efficacy and safety; NCT05980949)
Ongoing
ADEPT-4Phase 3Psychosis in Alzheimer’s Disease12-week, double-blind, placebo-controlled trial (acute efficacy; NCT06585787)Ongoing
TrialDevelopment phasePopulationTrial descriptionStatus
EMERGENT-1Phase 2Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT03697252)
Completed, results published
EMERGENT-2Phase 3Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT04659161)
Completed, results published
EMERGENT-3Phase 3Schizophrenia5-week, randomized, double-blind, placebo-controlled, inpatient trial
(acute efficacy and safety; NCT04738123)
Completed, results published
EMERGENT-4Phase 3Schizophrenia52-week, open-label extension trial of EMERGENT-2 and EMERGENT-3
(long-term safety; NCT04659174)
Completed
EMERGENT-5Phase 3Schizophrenia52-week, open-label trial
(long-term safety; NCT04820309)
Completed
ARISEPhase 3Schizophrenia6-week, randomized, double-blind, placebo-controlled trial of adjunctive xanomeline/trospium in people with inadequately controlled symptoms of schizophrenia (efficacy and safety of adjunctive xanomeline/trospium; NCT051453413)Ongoing
ARISE OLEPhase 3Schizophrenia52-week, outpatient, open-label extension trial of ARISE
(long-term safety and tolerability of adjunctive xanomeline/trospium; NCT05304767)
Ongoing
ADEPT-1Phase 3Psychosis in Alzheimer’s Disease12-week, single-blind treatment period followed by 26-week, double-blind,
placebo-controlled, randomized withdrawal period
(relapse prevention; NCT05511363)
Ongoing
ADEPT-2Phase 3Psychosis in Alzheimer’s Disease12-week, double-blind, placebo-controlled trial
(acute efficacy; NCT06126224)
Ongoing
ADEPT-3Phase 3Psychosis in Alzheimer’s Disease52-week, open-label extension trial of ADEPT-1 and ADEPT-2
(long-term efficacy and safety; NCT05980949)
Ongoing
ADEPT-4Phase 3Psychosis in Alzheimer’s Disease12-week, double-blind, placebo-controlled trial (acute efficacy; NCT06585787)Ongoing

Abbreviation: OLE, open-label extension.

The clinical development program for xanomeline/trospium included 3 pivotal, 5-week, randomized, double-blind, placebo-controlled trials in adults with schizophrenia experiencing an acute exacerbation of psychosis: EMERGENT-1 (NCT03697252; phase 2),104 EMERGENT-2 (NCT04659161; phase 3),105 and EMERGENT-3 (NCT04738123; phase 3).106 The 5-week EMERGENT trials were of similar design and enrolled adults with a primary diagnosis of schizophrenia based on the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, a PANSS total score of 80-120, a Clinical Global Impression-Severity (CGI-S) score ≥4, and recent worsening of psychosis warranting hospitalization. Participants were randomized 1:1 to twice-daily oral xanomeline/trospium or matched placebo for 5 weeks. Dosing started at xanomeline 50 mg/trospium chloride 20 mg and was titrated to a maximum dose of xanomeline 125 mg/trospium chloride 30 mg over 7 days. The primary endpoint in each trial was the change from baseline to week 5 in PANSS total score. Other outcome measures included change from baseline to week 5 in PANSS positive subscale, PANSS negative subscale, PANSS Marder negative factor, and CGI-S scale scores, as well as the proportion of participants achieving ≥30% reduction in PANSS total score. The long-term safety of xanomeline/trospium in schizophrenia was evaluated in the 52-week, open-label EMERGENT-4 (NCT04659174) and EMERGENT-5 (NCT04820309) trials. EMERGENT-4 was an extension trial enrolling participants who rolled over from EMERGENT-2 and EMERGENT-3, while EMERGENT-5 enrolled de novo participants who were not previously exposed to xanomeline/trospium. As previously discussed, xanomeline alone improved both psychotic symptoms and cognition in an early trial in people with AD.41 The ongoing ADEPT clinical development program is evaluating the potential of xanomeline/trospium for psychosis in AD, and the early trial supports further exploration of the potential of xanomeline/trospium for cognitive deficits in AD and related dementias, as well as potentially in other disorders with prominent cognitive deficits.

As xanomeline/trospium lacks D2 DA receptor activity and worked presynaptically to selectively decrease DA release in striatal regions outside of motor areas, there was great interest in the potential to augment antipsychotic response without incurring greater risk for D2-related motor or endocrine adverse effects. Preclinical data confirmed that xanomeline produced synergistic benefits in models of psychosis when coadministered with risperidone or the DA partial agonist aripiprazole.107 Given this rationale, the ongoing 5-week ARISE (NCT05145413) and 52-week ARISE open-label extension (NCT05304767) trials were designed to evaluate the efficacy, safety, and tolerability of xanomeline/trospium as adjunctive treatment in people whose symptoms of schizophrenia are inadequately controlled with current antipsychotic treatment.108

Finally, as discussed, in a phase 2 trial, xanomeline showed promise for its procognitive and antipsychotic effects in people with AD.41 The ongoing ADEPT clinical development program (ADEPT-1, NCT05511363; ADEPT-2, NCT06126224; ADEPT-3, NCT05980949; ADEPT-4, NCT06585787) is evaluating the efficacy, safety, and tolerability of xanomeline/trospium in the treatment of psychosis in people with AD in both acute and long-term trials.

EFFICACY OF XANOMELINE/TROSPIUM IN SCHIZOPHRENIA

In each of the acute EMERGENT trials, xanomeline/trospium significantly improved symptoms compared with placebo as demonstrated by the primary endpoint, the change from baseline to week 5 in PANSS total score. In pooled analyses of the acute EMERGENT trials, xanomeline/trospium was associated with a significantly greater 9.9-point reduction from baseline to week 5 in PANSS total score compared with placebo (Figure 5A; P < .0001; pooled Cohen’s d effect size, 0.65).109 Improvement in PANSS total score was significantly greater in the xanomeline/trospium vs placebo group starting at week 2, the earliest time point measured, and continuing through the end of the 5-week trials in each trial individually and in the pooled analysis.

Efficacy of xanomeline/trospium in the pooled 5-week acute EMERGENT trials.109 (A) PANSS total score. (B) CGI-S score. (C) PANSS positive subscale score. (D) PANSS negative subscale score. Values are LS mean ± SE. LS mean difference vs placebo: *P < .05; ****P < .0001. CGI-S, Clinical Global Impression-Severity; LS, least squares; PANSS, Positive and Negative Syndrome Scale; SEM, standard error of the mean.
Figure 5.

Efficacy of xanomeline/trospium in the pooled 5-week acute EMERGENT trials.109 (A) PANSS total score. (B) CGI-S score. (C) PANSS positive subscale score. (D) PANSS negative subscale score. Values are LS mean ± SE. LS mean difference vs placebo: *P < .05; ****P < .0001. CGI-S, Clinical Global Impression-Severity; LS, least squares; PANSS, Positive and Negative Syndrome Scale; SEM, standard error of the mean.

Improvements in PANSS total score with xanomeline/trospium were significant vs placebo across most participant subgroups assessed based on baseline demographics and characteristics, including age, race, gender, body mass index (BMI), and disease severity.110

Xanomeline/trospium was also associated with statistically significant and clinically meaningful reductions in PANSS positive and negative subscale scores. In pooled analyses of the acute EMERGENT trials, xanomeline/trospium was associated with significantly greater reductions compared with placebo at week 5 in PANSS positive subscale score (Figure 5C; −3.2 points; P < .0001; Cohen’s d effect size, 0.67), PANSS negative subscale score (Figure 5D; −1.7 points; P < .0001; Cohen’s d effect size, 0.40), and PANSS Marder negative factor score (−2.0 points P < .0001; Cohen’s d effect size, 0.42).109 These improvements were significantly greater with xanomeline/trospium vs placebo starting at week 2 for the PANSS positive subscale and at week 3 in PANSS negative subscale and PANSS Marder negative factor scores. In the pooled analyses, significance was maintained from the time of initial observation through week 5 across all primary and secondary efficacy measures.

To further evaluate the potential of benefits specifically on negative symptoms, a post hoc analysis111 was conducted to evaluate the effect of xanomeline/trospium on negative symptoms in a subgroup of the acute trial participants with moderate/severe negative symptoms and no predominance of positive symptoms at baseline based on the following previously published criteria112: (1) PANSS Marder negative factor score ≥ 24113; (2) PANSS Mohr positive score ≤ 19114; and (3) scores ≥ 4 on at least 2 of 3 PANSS blunted affect, passive/apathetic social withdrawal, or lack of spontaneity/flow of conversation items.115 Ten percent of the acute trial population (64/640) met these criteria. In this subgroup, xanomeline (n = 29) was associated with a significantly greater reduction from baseline to week 5 in PANSS Marder negative factor score compared with placebo (n = 35) (least squares mean difference = -4.71, P < .0001; Cohen’s d = 1.18). In addition, in both the full population and the subgroup with prominent negative symptoms, xanomeline/trospium was associated with significantly greater improvements from baseline to week 5 in the negative symptoms of both diminished emotional experience (Cohen’s d = 0.52 vs 1.08) and emotional expression (Cohen’s d = 0.22 vs 0.84). In the subgroup with prominent negative symptoms, the effect of xanomeline on negative symptoms remained significant after covarying for all types of symptoms, suggesting this treatment effect was not pseudospecific or secondary to improvement in other symptoms.

In the acute trials, responder analyses based on PANSS total and CGI-S score also favored xanomeline/trospium vs placebo.109 In the pooled analyses, a significantly greater proportion of participants achieved a ≥30% reduction in PANSS total score from baseline to week 5 in the xanomeline/trospium (41.4%) vs placebo (20.9%) groups. Similarly, 62.7% of participants in the xanomeline/trospium group experienced a ≥1-point improvement in CGI-S score at week 5 compared with 40.8% of participants in the placebo group.

The effect of xanomeline/trospium on cognitive impairment was evaluated as an exploratory endpoint in the 3 acute EMERGENT trials. EMERGENT-1 used the Cogstate Brief Battery,116 while EMERGENT-2 and EMERGENT-3 used the Cambridge Neuropsychological Test Automated Battery to assess cognitive function (Figure 6).117 The effect of xanomeline/trospium on cognitive performance was significant for those with baseline cognitive performance ≥1 SD below age-matched normative performance. In EMERGENT-1116 and pooled analyses for EMERGENT-2 and EMERGENT-3,117 the effect of xanomeline/trospium on cognitive performance was significant for those with baseline cognitive performance ≥1 SD below age-matched normative performance, which represented about 40% of participants across the 3 trials. Of note, this impact on cognition was not seen in the subgroup with milder baseline levels of cognitive dysfunction. Improvements in pooled analyses of EMERGENT-2/EMERGENT-3 were minimally correlated with changes in PANSS total, PANSS positive subscale, or PANSS negative subscale scores in both xanomeline/trospium and placebo treatment groups, suggesting any effect of xanomeline/trospium on cognition may not be pseudospecific and may be independent of changes in psychosis symptoms, although further research is needed.117

Effect of xanomeline/trospium on cognition (CANTAB composite score) in people with cognitive impairment at baseline in EMERGENT-2 and EMERGENT-3.117 Analysis in the subgroup of trial participants with cognitive impairment at baseline, defined as performing 1 SD below normal on the CANTAB. CANTAB, Cambridge Neuropsychological Test Automated Battery; LS, least squares; SE, standard error.
Figure 6.

Effect of xanomeline/trospium on cognition (CANTAB composite score) in people with cognitive impairment at baseline in EMERGENT-2 and EMERGENT-3.117 Analysis in the subgroup of trial participants with cognitive impairment at baseline, defined as performing 1 SD below normal on the CANTAB. CANTAB, Cambridge Neuropsychological Test Automated Battery; LS, least squares; SE, standard error.

The 52-week, open-label EMERGENT-4 and EMERGENT-5 trials were designed to provide long-term safety and tolerability data, while also examining changes in symptom response, bearing in mind the uncontrolled nature of the trial design. EMERGENT-4 was an open-label extension trial enrolling participants who completed the randomized, double-blind, placebo-controlled EMERGENT-2 or EMERGENT-3 trials and EMERGENT-5 enrolled participants with no prior exposure to xanomeline/trospium. An interim analysis of the EMERGENT-4 study (N = 110) noted that xanomeline/trospium treatment was associated with continuing improvement in PANSS total, PANSS positive and negative subscales, and PANSS Marder negative factor scores throughout the 52-week trial118 regardless of whether participants were previously treated with xanomeline/trospium or placebo during the acute trials.

SAFETY AND TOLERABILITY OF XANOMELINE/TROSPIUM IN SCHIZOPHRENIA

Overall Summary of Safety and Tolerability

In the 5-week, acute EMERGENT clinical trials, the majority of AEs with xanomeline/trospium related to muscarinic effects and were either procholinergic (eg, nausea, vomiting) or anticholinergic in nature (eg, dry mouth, constipation). Overall, these AEs were typically mild or moderate in intensity, tended to resolve with continued treatment, and were not associated with treatment discontinuation. There was no single AE leading to discontinuation that occurred at a rate ≥ 2% in participants treated with xanomeline/trospium and greater than the rate of placebo. Pooled data from the acute EMERGENT trials found the most common treatment-related AEs with an incidence of ≥2% in the xanomeline/trospium group and at a rate more than twice those in the placebo group were nausea (17% vs 3%), constipation (15% vs 5%), dyspepsia (12% vs 2%), vomiting (11% vs 1%), systemic hypertension (6% vs 1%), dry mouth (5% vs 2%), abdominal pain (5% vs 2%), tachycardia (5% vs 2%), dizziness (4% vs 2%), diarrhea (3% vs 2%), gastroesophageal reflux (3% vs <1%), and blurred vision (2% vs <1%) (Table 2).119 The discontinuation rates due to AEs were low and similar for the xanomeline/trospium and placebo groups (5.9% vs 4.4%).

Table 2.

Treatment-related AEs with xanomeline/trospium in the pooled 5-week acute EMERGENT trials.119

VariableXanomeline/
trospium
(n = 340)
Placebo
(n = 343)
Any treatment-related AE, n (%)176 (53)101 (29)
Serious treatment-related AE, n (%)1 (<1%)a0
Treatment-related AEs reported in ≥2% of people in the xanomeline/trospium group and at least twice the placebo rate, n (%)
 Nausea58 (17)11 (3)
 Constipation51 (15)18 (5)
 Dyspepsia41 (12)8 (2)
 Vomiting37 (11)3 (1)
 Hypertensionb20 (6)4 (1)
 Dry mouth17 (5)5 (2)
 Somnolence17 (5)14 (4)
 Tachycardia16 (5)7 (2)
 Abdominal pain16 (5)5 (2)
 Dizziness15 (4)6 (2)
 Gastroesophageal reflux disease9 (3)1 (<1)
 Vision blurred8 (2)1 (<1)
VariableXanomeline/
trospium
(n = 340)
Placebo
(n = 343)
Any treatment-related AE, n (%)176 (53)101 (29)
Serious treatment-related AE, n (%)1 (<1%)a0
Treatment-related AEs reported in ≥2% of people in the xanomeline/trospium group and at least twice the placebo rate, n (%)
 Nausea58 (17)11 (3)
 Constipation51 (15)18 (5)
 Dyspepsia41 (12)8 (2)
 Vomiting37 (11)3 (1)
 Hypertensionb20 (6)4 (1)
 Dry mouth17 (5)5 (2)
 Somnolence17 (5)14 (4)
 Tachycardia16 (5)7 (2)
 Abdominal pain16 (5)5 (2)
 Dizziness15 (4)6 (2)
 Gastroesophageal reflux disease9 (3)1 (<1)
 Vision blurred8 (2)1 (<1)

aPsychotic disorder (n = 1).

bHypertension is the MedDRA preferred term and is not necessarily reflective of clinical hypertension.

Abbreviations: AE, adverse event; MeDRA, Medical Dictionary for Medical Activities.

Table 2.

Treatment-related AEs with xanomeline/trospium in the pooled 5-week acute EMERGENT trials.119

VariableXanomeline/
trospium
(n = 340)
Placebo
(n = 343)
Any treatment-related AE, n (%)176 (53)101 (29)
Serious treatment-related AE, n (%)1 (<1%)a0
Treatment-related AEs reported in ≥2% of people in the xanomeline/trospium group and at least twice the placebo rate, n (%)
 Nausea58 (17)11 (3)
 Constipation51 (15)18 (5)
 Dyspepsia41 (12)8 (2)
 Vomiting37 (11)3 (1)
 Hypertensionb20 (6)4 (1)
 Dry mouth17 (5)5 (2)
 Somnolence17 (5)14 (4)
 Tachycardia16 (5)7 (2)
 Abdominal pain16 (5)5 (2)
 Dizziness15 (4)6 (2)
 Gastroesophageal reflux disease9 (3)1 (<1)
 Vision blurred8 (2)1 (<1)
VariableXanomeline/
trospium
(n = 340)
Placebo
(n = 343)
Any treatment-related AE, n (%)176 (53)101 (29)
Serious treatment-related AE, n (%)1 (<1%)a0
Treatment-related AEs reported in ≥2% of people in the xanomeline/trospium group and at least twice the placebo rate, n (%)
 Nausea58 (17)11 (3)
 Constipation51 (15)18 (5)
 Dyspepsia41 (12)8 (2)
 Vomiting37 (11)3 (1)
 Hypertensionb20 (6)4 (1)
 Dry mouth17 (5)5 (2)
 Somnolence17 (5)14 (4)
 Tachycardia16 (5)7 (2)
 Abdominal pain16 (5)5 (2)
 Dizziness15 (4)6 (2)
 Gastroesophageal reflux disease9 (3)1 (<1)
 Vision blurred8 (2)1 (<1)

aPsychotic disorder (n = 1).

bHypertension is the MedDRA preferred term and is not necessarily reflective of clinical hypertension.

Abbreviations: AE, adverse event; MeDRA, Medical Dictionary for Medical Activities.

In the 52-week, long-term, open-label trials, the tolerability and safety profile of xanomeline/trospium was consistent with that observed in the 5-week acute trials, with no new safety signals identified. In an interim pooled analysis from the open-label EMERGENT-4 and EMERGENT-5 trials, xanomeline/trospium was generally well tolerated in the outpatient setting across 52 weeks.120 The most common treatment-related AEs with an incidence of ≥2% in the xanomeline/trospium group were nausea (17%), vomiting (15%), constipation (14%), dry mouth (9%), dyspepsia (6%), dizziness (6%), hypertension (6%), diarrhea (5%), headache (4%), somnolence (4%), hyperhidrosis (3%), gastroesophageal reflux (2%), upper abdominal pain (2%), salivary hypersecretion (2%), and blurred vision (2%).

In both the acute and long-term trials, the AEs associated with xanomeline/trospium were consistent with the known side effects associated with muscarinic receptors, particularly in peripheral tissues. Importantly, across the EMERGENT clinical trial program, the rates of AEs commonly associated with D2 DA-binding antipsychotics (ie, motor symptoms, weight and metabolic changes, somnolence/sedation, hyperprolactinemia and related sexual AEs) were low and/or similar to placebo.

Motor Adverse Effects

In pooled analyses of the acute EMERGENT trials, few people treated with xanomeline/trospium (2%) or placebo (<1%) experienced treatment-related motor symptoms based on the preferred FDA terminology of “extrapyramidal adverse effects (EPS).” All EPS treatment-emergent adverse events (TEAEs) occurred at extremely low rates, were mild or moderate, and most resolved during the treatment period without dosage changes to trial medication. The most common treatment-related adverse motor symptom was akathisia, reported by 2 (<1%) and 1 (<1%) participants in the xanomeline/trospium and placebo groups, respectively. Treatment-related dystonia, dyskinesia, and extrapyramidal disorder were each reported only in a single participant in the xanomeline/trospium group and none in the placebo group. There were no reports of TD in the acute EMERGENT trials. Consistent with the lack of treatment-related motor AEs, there was no effect seen on the movement disorder scale scores in the 3 acute trials (Simpson-Angus Scale score: −0.1; Barnes Akathisia Rating Scale score: −0.1; Abnormal Involuntary Movement Scale score: 0)119 or the long-term trials (Simpson-Angus Scale score: −0.2; Barnes Akathisia Rating Scale score: 0; Abnormal Involuntary Movement Scale score: −0.1).120 Together, these results suggest that xanomeline/trospium is not associated with clinically meaningful motor symptoms as predicted by its pharmacology.

Weight Gain and Metabolic Effects

In both the acute and long-term EMERGENT trials, xanomeline/trospium was not generally associated with clinically meaningful weight gain or changes in metabolic parameters,119 both of which are common antipsychotic-associated AEs.121 It is worth noting that it is common for participants on placebo to experience weight gain in acute schizophrenia trials when such trials are conducted in inpatient settings. In pooled analyses across all 3 acute trials, exposure to xanomeline/trospium was associated with a 1.41 kg increase in body weight compared with a 1.94 kg increase in the placebo group. Approximately half as many people in the xanomeline/trospium group (5.3%) experienced clinically meaningful weight gain (defined as an increase of ≥7%) compared with placebo (11.4%), and approximately twice as many people in the xanomeline/trospium group (1.5%) lost ≥7% weight compared with placebo (0.7%). Similar results were observed for other weight-related factors such as BMI and waist circumference. In the 52-week open-label trials, there was a mean body weight change with xanomeline/trospium of −2.6 ± 0.5 kg over 52 weeks. After 52 weeks of treatment, the proportion of people with potentially clinically significant weight gain (≥7% increase; 4.6%) was lower than the proportion with potentially clinically significant weight loss (≥7% decrease; 17.6%). Similarly, the proportion of people with a TEAE of weight loss (3.9%) was higher than increased weight (1.8%). Xanomeline/trospium was not associated with clinically meaningful adverse changes in metabolic parameters (including HbA1c and lipid levels) when looking at either mean changes or threshold analysis.109 These data highlight the limited weight and metabolic impact of xanomeline/trospium, consistent with its muscarinic pharmacology and in contrast to that seen with many atypical antipsychotics.

Heart Rate and Blood Pressure

In addition to the EMERGENT trials, a dedicated 24-hour, open-label, inpatient ambulatory blood pressure (BP) monitoring trial in people living with schizophrenia was designed to definitively assess potential BP changes associated with xanomeline/trospium and to provide heart rate (HR) data. The trial was statistically powered to determine whether there was a clinically meaningful change in systolic BP, consistent with U.S. FDA guidance, and collected vital sign data 48 times per day to provide a 24-hour average change in vital sign parameters. In that trial, xanomeline/trospium was associated with a −0.6 mmHg change in systolic BP and was successful on the primary endpoint by determining statistically (P < .05) that xanomeline/trospium was not associated with a clinically meaningful increase in systolic BP (3 mmHg threshold established per U.S. FDA guidance). Xanomeline/trospium was associated with a small, non-significant increase in diastolic BP (+ 1.9 mmHg) and an increase in HR (+ 9.8 beats per minute [bpm]), which is consistent with the pooled data from both the acute and long-term trials.

In the acute and long-term EMERGENT trials, xanomeline/trospium was associated with small BP changes and an increase in HR, which peaked early during the first week of treatment and partially attenuated with continued treatment as assessed at 2 hours post dose, when plasma levels of xanomeline and trospium are both at peak levels.104-106 From baseline to week 5, mean systolic BP changed by + 0.7 mmHg (xanomeline/trospium) and 0 mmHg (placebo), and mean diastolic BP increased by 1.9 mmHg (xanomeline/trospium) and 0.4 mmHg (placebo). In the short-term placebo-controlled trials, change in HR from baseline to week 5 was 5.9 bpm greater for xanomeline/trospium than for placebo. In interim pooled analyses of the long-term trials, xanomeline/trospium was associated with increases in HR with peak elevations on day 14 (+ 7.7 bpm), which were partially attenuated with continued dosing; mean HR change from baseline to week 52 was + 1.7 bpm.

Prolactin

Xanomeline/trospium was not associated with clinically meaningful hyperprolactinemia or related sexual AEs. In the pooled acute trials, prolactin levels were similar between the xanomeline/trospium and placebo groups over the 5-week treatment period; mean change from baseline to week 5 was 0.8 ng/L vs −1.4 ng/L, respectively.119 Similarly, in the interim analysis of the long-term trials, xanomeline/trospium was not associated with significant changes in prolactin levels consistent with its muscarinic receptor pharmacology.120

SUMMARY

New treatments with novel mechanisms of action, broader efficacy, and better tolerability are needed to improve outcomes for people living with schizophrenia. Based on the serendipitous discovery that the dual M1/M4 muscarinic receptor agonist xanomeline could exhibit antipsychotic properties, a large body of preclinical and human evidence highlighted the potential for M1 and M4 muscarinic receptor activation as a promising potential treatment strategy for schizophrenia.26-28 The recent approval of xanomeline/trospium by the U.S. FDA in September 2024 represents the culmination of 3 decades of research on the role of muscarinic receptors in the pathophysiology of schizophrenia. Xanomeline is a dual M1/M4 preferring muscarinic receptor agonist without activity at D2 DA receptors and, as such, represents the first new class of medication for schizophrenia since antipsychotics were approved more than 70 years ago. Most chronic disorders, like schizophrenia, are heterogeneous and benefit from the availability of multiple classes of medications that can be used in place of or in conjunction with other treatment options. Due to its unique presynaptic mechanism of action and absence of D2 DA receptor binding, xanomeline/trospium opens up new opportunities for people with inadequate response to D2 DA receptor blockade.

In clinical trials, xanomeline/trospium significantly reduced schizophrenia symptoms vs placebo and was generally well tolerated. In addition to improvement in positive symptoms, xanomeline/trospium was associated with significantly greater improvement in negative symptoms, as well as in cognitive impairment in those participants with more severe impairment at baseline, compared with placebo. Analyses showed that improvements in negative symptoms and cognitive impairment with xanomeline/trospium were independent of psychosis symptoms reduction in all 3 short-term trials. In the context of acute psychosis trials, any improvement in negative and cognitive symptoms may be a product of pseudospecificity; however, analyses of these outcomes suggest that effects on those symptom domains were distinct and not a result of pseudospecific carryover related to overall symptom improvement. The most common AEs with xanomeline/trospium were consistent with the known activity of xanomeline and trospium at muscarinic receptors. Importantly, xanomeline/trospium was not associated with clinically significant motor symptoms, weight gain, hyperprolactinemia or related sexual side effects, or somnolence/sedation. Xanomeline/trospium is the first in a new class of non-D2 binding muscarinic receptor stimulating medications and, therefore, has the potential to be an important new treatment option for persons living with schizophrenia.

Acknowledgments

Medical writing and editorial support were provided by Matthew Jacobson, Isaac Dripps, PhD, and Paula Stuckart of Apollo Medical Communications, part of Helios Global Group, and funded by Bristol Myers Squibb.

Author contributions

Jonathan M. Meyer (Conceptualization [equal], Visualization [equal], Writing—original draft [equal], Writing—review & editing [equal]), Ken Kramer (Conceptualization [equal], Visualization [equal], Writing—original draft [equal], Writing—review & editing [equal]), Scott Vuocolo (Conceptualization [equal], Visualization [equal], Writing—original draft [equal], Writing—review & editing [equal]), Inder Kaul (Conceptualization [equal], Data curation [lead], Formal analysis [lead], Funding acquisition [equal], Investigation [lead], Methodology [lead], Project administration [lead], Resources [supporting], Supervision [lead], Validation [lead], Visualization [equal], Writing—original draft [equal], Writing—review & editing [equal]), and Andrew C. Miller (Conceptualization [equal], Data curation [equal], Formal analysis [supporting], Funding acquisition [equal], Investigation [supporting], Methodology [supporting], Project administration [equal], Resources [lead], Software [equal], Supervision [lead], Validation [supporting], Visualization [equal], Writing—original draft [equal], Writing—review & editing [equal])

Funding

None declared.

Conflicts of interest

In the prior 24 months, J.M.M. has received advising, consulting, and/or speaker fees from AbbVie, Alkermes, Axsome, BioXcel, Bristol Myers Squibb, Cerevel, Delpor, Intra-Cellular Therapies, Neurocrine, Noven, Otsuka America, Relmada, Sumitomo, and Teva. K.K., S.V., and I.K. are employees of Bristol Myers Squibb. A.C.M. is a consultant to Bristol Myers Squibb.

Data availability

No new data were generated or analyzed in support of this research.

References

1.

Marder
SR
,
Cannon
TD.
Schizophrenia
.
N Engl J Med.
2019
;
381
:
1753
1761
. https://doi.org/

2.

Jauhar
S
,
Johnstone
M
,
McKenna
PJ.
Schizophrenia
.
Lancet.
2022
;
399
:
473
486
. https://doi.org/

3.

Weye
N
,
Santomauro
DF
,
Agerbo
E
, et al.
Register-based metrics of years lived with disability associated with mental and substance use disorders: a register-based cohort study in Denmark
.
Lancet Psychiatry.
2021
;
8
:
310
319
. https://doi.org/

4.

GBD 2016 Disease and Injury Incidence and Prevalence Collaborators
.
Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016
.
Lancet.
2017
;
390
:
1211
1259
.

5.

Olfson
M
,
Gerhard
T
,
Huang
C
,
Crystal
S
,
Stroup
TS.
Premature mortality among adults with schizophrenia in the United States
.
JAMA Psychiatry.
2015
;
72
:
1172
1181
. https://doi.org/

6.

Saha
S
,
Chant
D
,
McGrath
J.
A systematic review of mortality in schizophrenia: is the differential mortality gap worsening over time
?
Arch Gen Psychiatry.
2007
;
64
:
1123
1131
. https://doi.org/

7.

Laursen
TM
,
Munk-Olsen
T
,
Gasse
C.
Chronic somatic comorbidity and excess mortality due to natural causes in persons with schizophrenia or bipolar affective disorder
.
PLoS One.
2011
;
6
:
e24597
. https://doi.org/

8.

Laursen
TM
,
Nordentoft
M
,
Mortensen
PB.
Excess early mortality in schizophrenia
.
Annu Rev Clin Psychol.
2014
;
10
:
425
448
. https://doi.org/

9.

Hjorthøj
C
,
Stürup
AE
,
McGrath
JJ
,
Nordentoft
M.
Years of potential life lost and life expectancy in schizophrenia: a systematic review and meta-analysis
.
Lancet Psychiatry.
2017
;
4
:
295
301
. https://doi.org/

10.

Stahl
S.
Stahl’s Essential Psychopharmacology.
5th ed.
Cambridge University Press
;
2021
.

11.

Correll
CU
,
Abi-Dargham
A
,
Howes
O.
Emerging treatments in schizophrenia
.
J Clin Psychiatry.
2022
;
83
:
SU21024IP21021
.

12.

Lobo
MC
,
Whitehurst
TS
,
Kaar
SJ
,
Howes
OD.
New and emerging treatments for schizophrenia: a narrative review of their pharmacology, efficacy and side effect profile relative to established antipsychotics
.
Neurosci Biobehav Rev.
2022
;
132
:
324
361
. https://doi.org/

13.

de Bartolomeis
A
,
Vellucci
L
,
Barone
A
, et al.
Clozapine’s multiple cellular mechanisms: What do we know after more than fifty years? A systematic review and critical assessment of translational mechanisms relevant for innovative strategies in treatment-resistant schizophrenia
.
Pharmacol Ther.
2022
;
236
:
108236
. https://doi.org/

14.

Kane
JM
,
Agid
O
,
Baldwin
ML
, et al.
Clinical guidance on the identification and management of treatment-resistant schizophrenia
.
J Clin Psychiatry.
2019
;
80
:
2783
.

15.

Joshi
YB.
Cholinergic functioning, cognition, and anticholinergic medication burden in schizophrenia
.
Curr Top Behav Neurosci.
2023
;
63
:
393
406
. https://doi.org/

16.

Kharkwal
G
,
Brami-Cherrier
K
,
Lizardi-Ortiz
JE
, et al.
Parkinsonism driven by antipsychotics originates from dopaminergic control of striatal cholinergic interneurons
.
Neuron.
2016
;
91
:
67
78
. https://doi.org/

17.

Huhn
M
,
Nikolakopoulou
A
,
Schneider-Thoma
J
, et al.
Comparative efficacy and tolerability of 32 oral antipsychotics for the acute treatment of adults with multi-episode schizophrenia: a systematic review and network meta-analysis
.
Lancet.
2019
;
394
:
939
951
. https://doi.org/

18.

Lacro
JP
,
Dunn
LB
,
Dolder
CR
,
Leckband
SG
,
Jeste
DV.
Prevalence of and risk factors for medication nonadherence in patients with schizophrenia: a comprehensive review of recent literature
.
J Clin Psychiatry.
2002
;
63
:
892
909
. https://doi.org/

19.

Pillinger
T
,
McCutcheon
RA
,
Vano
L
, et al.
Comparative effects of 18 antipsychotics on metabolic function in patients with schizophrenia, predictors of metabolic dysregulation, and association with psychopathology: a systematic review and network meta-analysis
.
Lancet Psychiatry.
2020
;
7
:
64
77
. https://doi.org/

20.

Lingjaerde
O.
Tetrabenazine (nitoman) in the treatment of psychoses. With a discussion on the central mode of action of tetrabenazine and reserpine
.
Acta Psychiatr Scand.
1963
;
39:Suppl170
:
171
109
.

21.

Kinon
BJ
,
Lieberman
JA.
Mechanisms of action of atypical antipsychotic drugs: a critical analysis
.
Psychopharmacology (Berl).
1996
;
124
:
2
34
. https://doi.org/

22.

Musco
S
,
McAllister
V
,
Caudle
I.
Dopamine-receptor blocking agent-associated akathisia: a summary of current understanding and proposal for a rational approach to treatment
.
Ther Adv Psychopharmacol.
2020
;
10
:
2045125320937575
. https://doi.org/

23.

McCutcheon
RA
,
Abi-Dargham
A
,
Howes
OD.
Schizophrenia, dopamine and the striatum: from biology to symptoms
.
Trends Neurosci.
2019
;
42
:
205
220
. https://doi.org/

24.

McCutcheon
R
,
Beck
K
,
Jauhar
S
,
Howes
OD.
Defining the locus of dopaminergic dysfunction in schizophrenia: a meta-analysis and test of the mesolimbic hypothesis
.
Schizophr Bull.
2018
;
44
:
1301
1311
. https://doi.org/

25.

Raedler
TJ
,
Bymaster
FP
,
Tandon
R
,
Copolov
D
,
Dean
B.
Towards a muscarinic hypothesis of schizophrenia
.
Mol Psychiatry.
2007
;
12
:
232
246
. https://doi.org/

26.

Paul
SM
,
Yohn
SE
,
Brannan
SK
,
Neugebaurer
NM
,
Breier
A.
Muscarinic receptor activators as novel treatments for schizophrenia
.
Biol Psych.
2024
;
96
:
627
637
.

27.

Paul
SM
,
Yohn
SE
,
Popiolek
M
,
Miller
AC
,
Felder
CC.
Muscarinic acetylcholine receptor agonists as novel treatments for schizophrenia
.
Am J Psychiatry.
2022
;
179
:
611
627
. https://doi.org/

28.

Yohn
SE
,
Weiden
PJ
,
Felder
CC
,
Stahl
SM.
Muscarinic acetylcholine receptors for psychotic disorders: bench-side to clinic
.
Trends Pharmacol Sci.
2022
;
43
:
1098
1112
. https://doi.org/

29.

Taylor
SF
,
Tso
IF.
GABA abnormalities in schizophrenia: a methodological review of in vivo studies
.
Schizophr Res.
2015
;
167
:
84
90
. https://doi.org/

30.

de Jonge
JC
,
Vinkers
CH
,
Hulshoff Pol
HE
,
Marsman
A.
GABAergic mechanisms in schizophrenia: linking postmortem and in vivo studies
.
Front Psychiatry.
2017
;
8
:
118
.

31.

Stahl
SM.
Beyond the dopamine hypothesis of schizophrenia to three neural networks of psychosis: dopamine, serotonin, and glutamate
.
CNS Spectr.
2018
;
23
:
187
191
. https://doi.org/

32.

Kruse
AO
,
Bustillo
JR.
Glutamatergic dysfunction in schizophrenia
.
Transl Psychiatry.
2022
;
12
:
500
. https://doi.org/

33.

McCutcheon
RA
,
Krystal
JH
,
Howes
OD.
Dopamine and glutamate in schizophrenia: biology, symptoms and treatment
.
World Psychiatry.
2020
;
19
:
15
33
. https://doi.org/

34.

Uno
Y
,
Coyle
JT.
Glutamate hypothesis in schizophrenia
.
Psychiatry Clin Neurosci.
2019
;
73
:
204
215
. https://doi.org/

35.

Ashcroft
GW
,
Macdougall
EJ
,
Barker
PA.
A comparison of tetrabenazine and chlorpromazine in chronic schizophrenia
.
J Ment Sci.
1961
;
107
:
287
293
. https://doi.org/

36.

Dean
B.
IUPHAR Review on muscarinic M1 and M4 receptors as drug treatment targets relevant to the molecular pathology of schizophrenia
.
Pharmacol Res.
2024
;
210
:
107510
. https://doi.org/

37.

Pfeiffer
CC
,
Jenney
EH.
The inhibition of the conditioned response and the counteraction of schizophrenia by muscarinic stimulation of the brain
.
Ann N Y Acad Sci.
1957
;
66
:
753
764
. https://doi.org/

38.

Sauerberg
P
,
Olesen
PH
,
Nielsen
S
, et al.
Novel functional M1 selective muscarinic agonists. Synthesis and structure-activity relationships of 3-(1,2,5-thiadiazolyl)-1,2,5,6-tetrahydro-1-methylpyridines
.
J Med Chem.
1992
;
35
:
2274
2283
. https://doi.org/

39.

Wood
MD
,
Murkitt
KL
,
Ho
M
, et al.
Functional comparison of muscarinic partial agonists at muscarinic receptor subtypes hM1, hM2, hM3, hM4 and hM5 using microphysiometry
.
Br J Pharmacol.
1999
;
126
:
1620
1624
. https://doi.org/

40.

Felder
CC
,
Goldsmith
PJ
,
Jackson
K
, et al.
Current status of muscarinic M1 and M4 receptors as drug targets for neurodegenerative diseases
.
Neuropharmacology.
2018
;
136
:
449
458
. https://doi.org/

41.

Bodick
NC
,
Offen
WW
,
Levey
AI
, et al.
Effects of xanomeline, a selective muscarinic receptor agonist, on cognitive function and behavioral symptoms in Alzheimer disease
.
Arch Neurol.
1997
;
54
:
465
473
. https://doi.org/

42.

Shekhar
A
,
Potter
WZ
,
Lightfoot
J
, et al.
Selective muscarinic receptor agonist xanomeline as a novel treatment approach for schizophrenia
.
Am J Psychiatry.
2008
;
165
:
1033
1039
. https://doi.org/

43.

McArthur
RA
,
Gray
J
,
Schreiber
R.
Cognitive effects of muscarinic M1 functional agonists in non-human primates and clinical trials
.
Curr Opin Investig Drugs.
2010
;
11
:
740
760
.

44.

Shannon
HE
,
Rasmussen
K
,
Bymaster
FP
, et al.
Xanomeline, an M(1)/M(4) preferring muscarinic cholinergic receptor agonist, produces antipsychotic-like activity in rats and mice
.
Schizophr Res.
2000
;
42
:
249
259
. https://doi.org/

45.

Stanhope
KJ
,
Mirza
NR
,
Bickerdike
MJ
, et al.
The muscarinic receptor agonist xanomeline has an antipsychotic-like profile in the rat
.
J Pharmacol Exp Ther.
2001
;
299
:
782
792
.

46.

Andersen
MB
,
Fink-Jensen
A
,
Peacock
L
, et al.
The muscarinic M1/M4 receptor agonist xanomeline exhibits antipsychotic-like activity in Cebus apella monkeys
.
Neuropsychopharmacology.
2003
;
28
:
1168
1175
. https://doi.org/

47.

Dencker
D
,
Wörtwein
G
,
Weikop
P
, et al.
Involvement of a subpopulation of neuronal M4 muscarinic acetylcholine receptors in the antipsychotic-like effects of the M1/M4 preferring muscarinic receptor agonist xanomeline
.
J Neurosci.
2011
;
31
:
5905
5908
. https://doi.org/

48.

Woolley
ML
,
Carter
HJ
,
Gartlon
JE
,
Watson
JM
,
Dawson
LA.
Attenuation of amphetamine-induced activity by the non-selective muscarinic receptor agonist, xanomeline, is absent in muscarinic M4 receptor knockout mice and attenuated in muscarinic M1 receptor knockout mice
.
Eur J Pharmacol.
2009
;
603
:
147
149
. https://doi.org/

49.

Barak
S
,
Weiner
I.
The M₁/M₄ preferring agonist xanomeline reverses amphetamine-, MK801- and scopolamine-induced abnormalities of latent inhibition: putative efficacy against positive, negative and cognitive symptoms in schizophrenia
.
Int J Neuropsychopharmacol.
2011
;
14
:
1233
1246
. https://doi.org/

50.

Yohn
SE
,
Conn
PJ.
Positive allosteric modulation of M(1) and M(4) muscarinic receptors as potential therapeutic treatments for schizophrenia
.
Neuropharmacology.
2018
;
136
:
438
448
. https://doi.org/

51.

Yohn
SE
,
Harvey
PD
,
Brannan
SK
,
Horan
WP.
The potential of muscarinic M1 and M4 activators for the treatment of cognitive impairment with schizophrenia
.
Front Psychiatry.
2024
;
15
:
1421554
.

52.

Ford
CP.
The role of D2-autoreceptors in regulating dopamine neuron activity and transmission
.
Neuroscience.
2014
;
282
:
13
22
. https://doi.org/

53.

Scarr
E
,
Sundram
S
,
Keriakous
D
,
Dean
B.
Altered hippocampal muscarinic M4, but not M1, receptor expression from subjects with schizophrenia
.
Biol Psychiatry.
2007
;
61
:
1161
1170
. https://doi.org/

54.

Crook
JM
,
Tomaskovic-Crook
E
,
Copolov
DL
,
Dean
B.
Decreased muscarinic receptor binding in subjects with schizophrenia: a study of the human hippocampal formation
.
Biol Psychiatry.
2000
;
48
:
381
388
. https://doi.org/

55.

Scarr
E
,
Um
JY
,
Cowie
TF
,
Dean
B.
Cholinergic muscarinic M4 receptor gene polymorphisms: a potential risk factor and pharmacogenomic marker for schizophrenia
.
Schizophr Res.
2013
;
146
:
279
284
. https://doi.org/

56.

Scarr
E
,
Craig
JM
,
Cairns
MJ
, et al.
Decreased cortical muscarinic M1 receptors in schizophrenia are associated with changes in gene promoter methylation, mRNA and gene targeting microRNA
.
Transl Psychiatry.
2013
;
3
:
e230
. https://doi.org/

57.

Perry
E
,
Walker
M
,
Grace
J
,
Perry
R.
Acetylcholine in mind: a neurotransmitter correlate of consciousness
?
Trends Neurosci.
1999
;
22
:
273
280
. https://doi.org/

58.

Gerber
DJ
,
Sotnikova
TD
,
Gainetdinov
RR
, et al.
Hyperactivity, elevated dopaminergic transmission, and response to amphetamine in M1 muscarinic acetylcholine receptor-deficient mice
.
Proc Natl Acad Sci U S A.
2001
;
98
:
15312
15317
. https://doi.org/

59.

Gomeza
J
,
Zhang
L
,
Kostenis
E
, et al.
Enhancement of D1 dopamine receptor-mediated locomotor stimulation in M(4) muscarinic acetylcholine receptor knockout mice
.
Proc Natl Acad Sci U S A.
1999
;
96
:
10483
10488
. https://doi.org/

60.

Koshimizu
H
,
Leiter
LM
,
Miyakawa
T.
M4 muscarinic receptor knockout mice display abnormal social behavior and decreased prepulse inhibition
.
Mol Brain.
2012
;
5
:
10
. https://doi.org/

61.

Pozhidaev
IV
,
Boiko
AS
,
Loonen
AJM
, et al.
Association of cholinergic muscarinic M4 receptor gene polymorphism with schizophrenia
.
Appl Clin Genet.
2020
;
13
:
97
105
. https://doi.org/

62.

Scarr
E
,
Sundram
S
,
Deljo
A
, et al.
Muscarinic M1 receptor sequence: preliminary studies on its effects on cognition and expression
.
Schizophr Res.
2012
;
138
:
94
98
. https://doi.org/

63.

Cropley
VL
,
Scarr
E
,
Fornito
A
, et al.
The effect of a muscarinic receptor 1 gene variant on grey matter volume in schizophrenia
.
Psychiatry Res.
2015
;
234
:
182
187
. https://doi.org/

64.

Dean
B
,
McLeod
M
,
Keriakous
D
,
McKenzie
J
,
Scarr
E.
Decreased muscarinic1 receptors in the dorsolateral prefrontal cortex of subjects with schizophrenia
.
Mol Psychiatry.
2002
;
7
:
1083
1091
. https://doi.org/

65.

Raedler
TJ
,
Knable
MB
,
Jones
DW
, et al.
In vivo determination of muscarinic acetylcholine receptor availability in schizophrenia
.
Am J Psychiatry.
2003
;
160
:
118
127
. https://doi.org/

66.

Dean
B
,
Scarr
E.
Muscarinic M1 and M4 receptors: hypothesis driven drug development for schizophrenia
.
Psychiatry Res.
2020
;
288
:
112989
. https://doi.org/

67.

Scarr
E
,
Cowie
TF
,
Kanellakis
S
, et al.
Decreased cortical muscarinic receptors define a subgroup of subjects with schizophrenia
.
Mol Psychiatry.
2009
;
14
:
1017
1023
. https://doi.org/

68.

Tzavara
ET
,
Bymaster
FP
,
Davis
RJ
, et al.
M4 muscarinic receptors regulate the dynamics of cholinergic and dopaminergic neurotransmission: relevance to the pathophysiology and treatment of related CNS pathologies
.
FASEB J.
2004
;
18
:
1410
1412
. https://doi.org/

69.

Buck
SA
,
Torregrossa
MM
,
Logan
RW
,
Freyberg
Z.
Roles of dopamine and glutamate co-release in the nucleus accumbens in mediating the actions of drugs of abuse
.
FEBS J.
2021
;
288
:
1462
1474
. https://doi.org/

70.

Weiden
PJ
,
Yohn
SE
,
Felder
CC.
Acetylcholine as a regulator of dopamine pathways:
rationale for selective muscarinic agonists as candidates for antipsychotic drug development
. Presented at 2022 Annual Conference of the Schizophrenia International Research Society (SIRS). Florence, Italy; 2022.

71.

Yi
F
,
Ball
J
,
Stoll
KE
, et al.
Direct excitation of parvalbumin-positive interneurons by M1 muscarinic acetylcholine receptors: roles in cellular excitability, inhibitory transmission and cognition
.
J Physiol.
2014
;
592
:
3463
3494
. https://doi.org/

72.

Tikhonova
TB
,
Miyamae
T
,
Gulchina
Y
,
Lewis
DA
,
Gonzalez-Burgos
G.
Cell type- and layer-specific muscarinic potentiation of excitatory synaptic drive onto parvalbumin neurons in mouse prefrontal cortex
.
eNeuro
.
2018
;
5
:
ENEURO.0208
ENEU18.2018
. https://doi.org/

73.

Salgado
S
,
Kaplitt
MG.
The nucleus accumbens: a comprehensive review
.
Stereotact Funct Neurosurg.
2015
;
93
:
75
93
. https://doi.org/

74.

Patel
KR
,
Cherian
J
,
Gohil
K
,
Atkinson
D.
Schizophrenia: overview and treatment options
.
P T.
2014
;
39
:
638
645
.

75.

Forster
GL
,
Blaha
CD.
Pedunculopontine tegmental stimulation evokes striatal dopamine efflux by activation of acetylcholine and glutamate receptors in the midbrain and pons of the rat
.
Eur J Neurosci.
2003
;
17
:
751
762
. https://doi.org/

76.

Scarr
E
,
Hopper
S
,
Vos
V
, et al.
Low levels of muscarinic M1 receptor-positive neurons in cortical layers III and V in Brodmann areas 9 and 17 from individuals with schizophrenia
.
J Psychiatry Neurosci.
2018
;
43
:
338
346
. https://doi.org/

77.

Reddy-Thootkur
M
,
Kraguljac
NV
,
Lahti
AC.
The role of glutamate and GABA in cognitive dysfunction in schizophrenia and mood disorders - a systematic review of magnetic resonance spectroscopy studies
.
Schizophr Res.
2022
;
249
:
74
84
. https://doi.org/

78.

Spurny
B
,
Seiger
R
,
Moser
P
, et al.
Hippocampal GABA levels correlate with retrieval performance in an associative learning paradigm
.
Neuroimage.
2020
;
204
:
116244
. https://doi.org/

79.

Tamminga
CA
,
Southcott
S
,
Sacco
C
,
Wagner
AD
,
Ghose
S.
Glutamate dysfunction in hippocampus: relevance of dentate gyrus and CA3 signaling
.
Schizophr Bull.
2012
;
38
:
927
935
. https://doi.org/

80.

Dasari
S
,
Gulledge
AT.
M1 and M4 receptors modulate hippocampal pyramidal neurons
.
J Neurophysiol.
2011
;
105
:
779
792
. https://doi.org/

81.

Oda
S
,
Tsuneoka
Y
,
Yoshida
S
, et al.
Immunolocalization of muscarinic M1 receptor in the rat medial prefrontal cortex
.
J Comp Neurol.
2018
;
526
:
1329
1350
. https://doi.org/

82.

Ma
XH
,
Zhong
P
,
Gu
Z
,
Feng
J
,
Yan
Z.
Muscarinic potentiation of GABA(A) receptor currents is gated by insulin signaling in the prefrontal cortex
.
J Neurosci.
2003
;
23
:
1159
1168
. https://doi.org/

83.

Shirey
JK
,
Brady
AE
,
Jones
PJ
, et al.
A selective allosteric potentiator of the M1 muscarinic acetylcholine receptor increases activity of medial prefrontal cortical neurons and restores impairments in reversal learning
.
J Neurosci.
2009
;
29
:
14271
14286
. https://doi.org/

84.

Shirey
JK
,
Xiang
Z
,
Orton
D
, et al.
An allosteric potentiator of M4 mAChR modulates hippocampal synaptic transmission
.
Nat Chem Biol.
2008
;
4
:
42
50
. https://doi.org/

85.

Bubser
M
,
Bridges
TM
,
Dencker
D
, et al.
Selective activation of M4 muscarinic acetylcholine receptors reverses MK-801-induced behavioral impairments and enhances associative learning in rodents
.
ACS Chem Neurosci.
2014
;
5
:
920
942
. https://doi.org/

86.

Galloway
CR
,
Lebois
EP
,
Shagarabi
SL
,
Hernandez
NA
,
Manns
JR.
Effects of selective activation of M1 and M4 muscarinic receptors on object recognition memory performance in rats
.
Pharmacology.
2014
;
93
:
57
64
. https://doi.org/

87.

Gould
RW
,
Grannan
MD
,
Gunter
BW
, et al.
Cognitive enhancement and antipsychotic-like activity following repeated dosing with the selective M(4) PAM VU0467154
.
Neuropharmacology.
2018
;
128
:
492
502
. https://doi.org/

88.

Wu
L
,
D
.
C
,
Jia
H
,
Zhao
L
,
Liu
N
,
Shen
H.
NS-136, a selective muscarinic receptor positive allosteric modulator (PAM) for the treatment of schizophrenia
. Presented at the 2023 Society for Neuroscience Annual Meeting. Washington, DC; 2023.
Poster 242.209
.

89.

Lange
HS
,
Vardigan
JD
,
Cannon
CE
, et al.
Effects of a novel M4 muscarinic positive allosteric modulator on behavior and cognitive deficits relevant to Alzheimer’s disease and schizophrenia in rhesus monkey
.
Neuropharmacology.
2021
;
197
:
108754
. https://doi.org/

90.

Byun
NE
,
Grannan
M
,
Bubser
M
, et al.
Antipsychotic drug-like effects of the selective M4 muscarinic acetylcholine receptor positive allosteric modulator VU0152100
.
Neuropsychopharmacology.
2014
;
39
:
1578
1593
. https://doi.org/

91.

Wilson
H
,
de Natale
ER
,
Politis
M.
Nucleus basalis of Meynert degeneration predicts cognitive impairment in Parkinson’s disease
.
Handb Clin Neurol.
2021
;
179
:
189
205
. https://doi.org/

92.

Anagnostaras
SG
,
Murphy
GG
,
Hamilton
SE
, et al.
Selective cognitive dysfunction in acetylcholine M1 muscarinic receptor mutant mice
.
Nat Neurosci.
2003
;
6
:
51
58
. https://doi.org/

93.

Drinkenburg
WH
,
Sondag
HN
,
Coenders
CJ
,
Andrews
JS
,
Vossen
JM.
Effects of selective antagonism or depletion of the cholinergic system on visual discrimination performance in rats
.
Behav Pharmacol.
1995
;
6
:
695
702
.

94.

Hunter
AJ
,
Roberts
FF.
The effect of pirenzepine on spatial learning in the Morris Water Maze
.
Pharmacol Biochem Behav.
1988
;
30
:
519
523
. https://doi.org/

95.

Gould
RW
,
Dencker
D
,
Grannan
M
, et al.
Role for the M1 muscarinic acetylcholine receptor in top-down cognitive processing using a touchscreen visual discrimination task in mice
.
ACS Chem Neurosci.
2015
;
6
:
1683
1695
. https://doi.org/

96.

Thorn
CA
,
Popiolek
M
,
Stark
E
,
Edgerton
JR.
Effects of M1 and M4 activation on excitatory synaptic transmission in CA1
.
Hippocampus.
2017
;
27
:
794
810
. https://doi.org/

97.

Ghoshal
A
,
Rook
JM
,
Dickerson
JW
, et al.
Potentiation of M1 muscarinic receptor reverses plasticity deficits and negative and cognitive symptoms in a schizophrenia mouse model
.
Neuropsychopharmacology.
2016
;
41
:
598
610
. https://doi.org/

98.

Kurimoto
E
,
Yamada
R
,
Hirakawa
T
,
Kimura
H.
Therapeutic potential of TAK-071, a muscarinic M(1) receptor positive allosteric modulator with low cooperativity, for the treatment of cognitive deficits and negative symptoms associated with schizophrenia
.
Neurosci Lett.
2021
;
764
:
136240
. https://doi.org/

99.

Bubser
M
,
Byun
N
,
Wood
MR
,
Jones
CK.
Muscarinic receptor pharmacology and circuitry for the modulation of cognition
. Handb Exp Pharmacol. 2012;(208):121–166.  https://doi.org/

100.

Hassani
SA
,
Neumann
A
,
Russell
J
,
Jones
CK
,
Womelsdorf
T.
M(1)-selective muscarinic allosteric modulation enhances cognitive flexibility and effective salience in nonhuman primates
.
Proc Natl Acad Sci U S A.
2023
;
120
:
e2216792120
. https://doi.org/

101.

Todorova
A
,
Vonderheid-Guth
B
,
Dimpfel
W.
Effects of tolterodine, trospium chloride, and oxybutynin on the central nervous system
.
J Clin Pharmacol.
2001
;
41
:
636
644
. https://doi.org/

102.

Staskin
D
,
Kay
G
,
Tannenbaum
C
, et al.
Trospium chloride is undetectable in the older human central nervous system
.
J Am Geriatr Soc.
2010
;
58
:
1618
1619
. https://doi.org/

103.

Kavoussi
R
,
Miller
AC
,
Breier
A.
Xanomeline plus trospium: a novel strategy to enhance pro-muscarinic efficacy and mitigate peripheral side effects
. Presented at the 2017 American Society for Clinical Psychopharmacology (ASCP) Annual Meeting. Miami, Florida; 2017.

104.

Brannan
SK
,
Sawchak
S
,
Miller
AC
, et al.
Muscarinic cholinergic receptor agonist and peripheral antagonist for schizophrenia
.
N Engl J Med.
2021
;
384
:
717
726
. https://doi.org/

105.

Kaul
I
,
Sawchak
S
,
Correll
CU
, et al.
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
. https://doi.org/

106.

Kaul
I
,
Sawchak
S
,
Walling
DP
, et al.
Efficacy and safety of xanomeline-trospium chloride in schizophrenia: a randomized clinical trial
.
JAMA Psychiatry.
2024
;
81
:
749
756
. https://doi.org/

107.

McKinzie
DL
,
Felder
CC
,
Fischer
K
, et al.
The muscarinic agnist xanomeline demonstrates standalone activity and augments clinical antipsychotics in rodent behavioral models of pychosis
. Presented at the Schizophrenia International Research Society (SIRS) 2022 Annual Congress. Florence, Italy; 2022.

108.

Kinon
BJ
,
Leucht
S
,
Tamminga
C
, et al.
Rationale for adjunctive treatment targeting multiple mechanisms in schizophrenia
.
J Clin Psychiatry.
2024
;
85
:
23nr15240
. https://doi.org/

109.

Kaul
I
,
Citrome
L
,
Sawchak
S
, et al.
Efficacy of KarXT (xanomeline-trospium) in schizophrenia: pooled results from the randomized, double-blind, placebo-controlled EMERGENT trials
. Presented at the 62nd Annual Meeting of the American College of Neuropsychopharmacology (ACNP). Tampa, Florida; 2023.
Poster M74
.

110.

Kaul
I
,
Correll
CU
,
Sawchak
S
,
Miller
AC
,
Paul
SM
,
Brannan
SK.
KarXT (xanomeline-trospium) demonstrates broad efficacy in people with schizophrenia across a wide range of demographic subgroups: pooled results from the 3 randomized, double-blind, placebo-controlled EMERGENT trials
. Presented at the 62nd Annual Meeting of the American College of Neuropsychopharmacology (ACNP). Tampa, Florida; 2023.
Poster T186
.

111.

Horan
WP
,
Targum
SD
,
Claxton
A
, et al.
Efficacy of KarXT on negative symptoms in acute schizophrenia: a post hoc analysis of pooled data from 3 trials
.
Schizophr Res.
2024
;
274
:
57
65
. https://doi.org/

112.

Earley
W
,
Guo
H
,
Daniel
D
, et al.
Efficacy of cariprazine on negative symptoms in patients with acute schizophrenia: a post hoc analysis of pooled data
.
Schizophr Res.
2019
;
204
:
282
288
. https://doi.org/

113.

Marder
SR
,
Davis
JM
,
Chouinard
G.
The effects of risperidone on the five dimensions of schizophrenia derived by factor analysis: combined results of the North American trials
.
J Clin Psychiatry.
1997
;
58
:
538
546
. https://doi.org/

114.

Mohr
PE
,
Cheng
CM
,
Claxton
K
, et al.
The heterogeneity of schizophrenia in disease states
.
Schizophr Res.
2004
;
71
:
83
95
. https://doi.org/

115.

Kay
SR
,
Fiszbein
A
,
Opler
LA.
The positive and negative syndrome scale (PANSS) for schizophrenia
.
Schizophr Bull.
1987
;
13
:
261
276
. https://doi.org/

116.

Sauder
C
,
Allen
LA
,
Baker
E
, et al.
Effectiveness of KarXT (xanomeline-trospium) for cognitive impairment in schizophrenia: post hoc analyses from a randomised, double-blind, placebo-controlled phase 2 study
.
Transl Psychiatry.
2022
;
12
:
491
. https://doi.org/

117.

Horan
WP
,
Sauder
C
,
Harvey
PD
, et al.
The impact of KarXT on cognitive impairment in acute schizophrenia: replication in pooled data from two phase 3 trials
.
Am J Psychiatry.
2025;182(3):277–306.

118.

Kaul
I
,
Cohen
EA
,
Miller
AC
,
Claxton
A
,
Paul
SM
,
Brannan
SK.
Maintenance of efficacy of KarXT (xanomeline and trospium) in schizophrenia
. Presented at 2024 Annual Conference of the Schizophrenia International Research Society (SIRS). Florence, Italy; 2024.
Poster F264
.

119.

Brannan
SK
,
Cutler
AJ
,
Sawchak
S
, et al.
Safety and tolerability of KarXT (xanomeline-trospium): pooled results from the randomized, double-blind, placebo-controlled EMERGENT trials
. Presented at the 62nd Annual Meeting of the American College of Neuropsychopharmacology (ACNP). Tampa, Florida; 2023.
Poster T189
.

120.

Marcus
R
,
Kakar
R
,
Miller
AC
, et al.
Long-term safety of KarXT (xanomeline and trospium) in schizophrenia
. Presented at 2024 Annual Conference of the Schizophrenia International Research Society (SIRS). Florence, Italy; 2023.
Poster F74
.

121.

Barton
BB
,
Segger
F
,
Fischer
K
,
Obermeier
M
,
Musil
R.
Update on weight-gain caused by antipsychotics: a systematic review and meta-analysis
.
Expert Opin Drug Saf.
2020
;
19
:
295
314
. https://doi.org/

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.