AZD-7325 / BAER-101: A Comprehensive Monograph on a Subtype-Selective GABAA Modulator
Executive Summary
AZD-7325 is an investigational small molecule that represents a significant endeavor in the rational design of central nervous system (CNS) therapeutics. Developed initially by AstraZeneca, it is a novel, orally active positive allosteric modulator (PAM) of the γ-aminobutyric acid type A (GABAA) receptor. Its design was predicated on a refined understanding of GABAA receptor neuropharmacology, specifically engineered for high affinity and functional selectivity for receptor subtypes containing α2 and α3 subunits, while demonstrating minimal activity at the α1 and α5 subunits. This profile was hypothesized to confer potent anxiolytic and anticonvulsant effects without the dose-limiting side effects of traditional non-selective benzodiazepines, such as sedation, cognitive impairment, and abuse liability, which are primarily mediated by the α1 and α5 subunits, respectively.
Preclinical studies validated this hypothesis, demonstrating potent anxiolytic-like effects and, more notably, profound anti-seizure activity in robust animal models of epilepsy and Fragile X Syndrome, all with a favorable safety margin. Early-phase clinical trials in healthy volunteers further confirmed its differentiated profile, showing high levels of GABAA receptor occupancy in the brain without inducing the significant CNS impairment characteristic of drugs like lorazepam. Despite this promising profile, AZD-7325 failed to demonstrate statistically significant efficacy over placebo in two large Phase II proof-of-concept trials for Generalized Anxiety Disorder (GAD), its primary target indication. This outcome led AstraZeneca to discontinue its development.
However, the extensive and high-quality data package generated by AstraZeneca, which established a clear safety profile in over 700 subjects and highlighted strong signals in disorders of neuronal hyperexcitability, made the compound an attractive asset for out-licensing. In 2019, the molecule was acquired by Baergic Bio, a partner company of Fortress Biotech, and renamed BAER-101. Its development was subsequently reoriented towards indications more closely aligned with its demonstrated biological strengths. The current strategy, under Avenue Therapeutics, focuses on epilepsy and other CNS disorders characterized by neuronal hyperexcitability. The journey of AZD-7325 to BAER-101 serves as a compelling case study in modern pharmaceutical R&D, illustrating the principles of asset-centric biotech strategy, the challenges of translating refined pharmacology into clinical efficacy, and the potential for data-driven repurposing of scientifically sound but commercially deprioritized molecules. This report provides a definitive and exhaustive analysis of its chemical properties, pharmacological profile, complex clinical history, and the scientific rationale underpinning its future development.
Section 1: Chemical Identity and Physicochemical Properties
A precise and unambiguous definition of a molecule's chemical and physical characteristics is fundamental to any comprehensive analysis. This section details the nomenclature, structural identifiers, and key physicochemical properties of AZD-7325, establishing a foundational reference for the compound.
1.1 Nomenclature and Identifiers
The compound has been known by several names and is cataloged across numerous chemical and biomedical databases under various identifiers.
- Primary Development Name: The compound was originally developed and clinically tested by AstraZeneca under the designation AZD-7325.[1] It is also commonly referenced without the hyphen as AZD7325 or with a space as AZD 7325.[1]
- Redevelopment Name: Following its out-licensing from AstraZeneca, the compound was renamed BAER-101 for its continued development.[5]
- Systematic (IUPAC) Name: The formal chemical name according to the International Union of Pure and Applied Chemistry (IUPAC) nomenclature is 4-amino-8-(2-fluoro-6-methoxyphenyl)-N-propylcinnoline-3-carboxamide.[1]
- CAS Registry Number: The unique identifier assigned by the Chemical Abstracts Service is 942437-37-8.[1]
- DrugBank Accession Number: The compound is cataloged in the DrugBank database under the accession number DB13994.[1]
- Other Identifiers: To facilitate cross-referencing across different research platforms, a list of additional key identifiers is provided in Table 1. These include its Unique Ingredient Identifier (UNII), European Community (EC) Number, and ChEMBL ID.[1]
1.2 Chemical Structure and Molecular Formula
AZD-7325 is a synthetic organic compound belonging to the cinnoline class of heterocyclic molecules.[2]
- Molecular Formula: The empirical formula for AZD-7325 is $C_{19}H_{19}FN_{4}O_{2}$.[1]
- Chemical Structure: The molecule is built upon a 4-aminocinnoline-3-carboxamide core. An N-propyl group is attached to the carboxamide nitrogen, and an 8-position aryl substitution is present, specifically a 2-fluoro-6-methoxyphenyl group.[2] This cinnoline moiety was a key scaffold in AstraZeneca's medicinal chemistry program and was amenable to radiolabeling for metabolic studies.[2]
- Structural Line Notations: For computational chemistry and database searching, the structure is represented by the following standard line notations:
- SMILES: CCNC(=O)C1=NN=C2C(=C1N)C=CC=C2C3=C(C=CC=C3F)OC.[1]
- InChI: InChI=1S/C19H19FN4O2/c1-3-10-22-19(25)18-16(21)12-7-4-6-11(17(12)23-24-18)15-13(20)8-5-9-14(15)26-2/h4-9H,3,10H2,1-2H3,(H2,21,23)(H,22,25).[1]
- InChIKey: KYDURMHFWXCKMW-UHFFFAOYSA-N.[1]
1.3 Physicochemical Properties
The physicochemical properties of AZD-7325 are consistent with those of an orally bioavailable small molecule drug intended for CNS penetration.
- Molecular Weight: The calculated molecular weight is approximately 354.38 g/mol to 354.4 g/mol.[1] The monoisotopic mass is computed as 354.14920402 Da.[1]
- Solubility: The compound is reported to be soluble in dimethyl sulfoxide (DMSO), a common solvent for in vitro biological assays.[3] A standard concentration for stock solutions is 10 mM in DMSO.[9]
- Storage and Stability: As a solid powder, AZD-7325 is stable for at least four years when stored at -20°C.[3] In solution, storage at -80°C is recommended for periods up to six months.[9]
- Drug-Likeness and Lipophilicity: The molecule exhibits properties that predict good oral absorption and membrane permeability. It adheres to Lipinski's Rule-of-Five, with zero violations reported.[4] Key computed parameters include:
- Hydrogen Bond Acceptors: 5.[1]
- Hydrogen Bond Donors: 2.[1]
- Rotatable Bonds: 6.[1]
- Topological Polar Surface Area (TPSA): 90.1 Ų.[1]
- LogP (Octanol-Water Partition Coefficient): The computed partition coefficient (XLogP3) is approximately 3.2 to 3.3, indicating moderate lipophilicity suitable for crossing the blood-brain barrier.[1]
A consolidated summary of these key identifiers and properties is presented in Table 1.
Table 1: Key Identifiers and Physicochemical Properties of AZD-7325
| Property | Value | Source(s) |
|---|
| Primary Names | AZD-7325, BAER-101 | 1 |
| IUPAC Name | 4-amino-8-(2-fluoro-6-methoxyphenyl)-N-propylcinnoline-3-carboxamide | 1 |
| CAS Number | 942437-37-8 | 1 |
| DrugBank ID | DB13994 | 1 |
| UNII | KNM216XOUH | 1 |
| ChEMBL ID | CHEMBL1783282 | 1 |
| Molecular Formula | $C_{19}H_{19}FN_{4}O_{2}$ | 1 |
| Molecular Weight | 354.4 g/mol | 1 |
| SMILES | CCNC(=O)C1=NN=C2C(=C1N)C=CC=C2C3=C(C=CC=C3F)OC | 1 |
| InChIKey | KYDURMHFWXCKMW-UHFFFAOYSA-N | 1 |
| Solubility | Soluble in DMSO | 3 |
| Storage (Solid) | -20°C (≥ 4 years stability) | 3 |
| XLogP | 3.2 - 3.3 | 1 |
| H-Bond Donors | 2 | 1 |
| H-Bond Acceptors | 5 | 1 |
| Lipinski's Rules Broken | 0 | 4 |
Section 2: Nonclinical Pharmacology and Toxicology
The therapeutic rationale for AZD-7325 is rooted in a sophisticated understanding of the GABAA receptor system. This section details the fundamental neuropharmacology of GABAA receptor subtypes, elucidates the specific mechanism of action of AZD-7325, and summarizes its pharmacokinetic, metabolic, preclinical efficacy, and safety profile.
2.1 The GABAA Receptor System: A Primer on Subtype-Selective Modulation
The balance between neuronal excitation and inhibition is critical for all CNS functions, with γ-aminobutyric acid (GABA) serving as the principal inhibitory neurotransmitter in the mammalian brain.[11] GABA exerts its primary effects through GABAA receptors, which are ligand-gated ion channels belonging to the Cys-loop superfamily.[11] These receptors are heteropentameric complexes, typically composed of two α, two β, and one γ subunit, which assemble to form a central chloride-permeable pore.[2] Upon binding of GABA, the channel opens, allowing chloride ions to flow into the neuron, which hyperpolarizes the cell membrane and reduces the probability of firing an action potential, thus mediating fast synaptic inhibition.[11]
The immense diversity of the GABAA receptor system, arising from 19 different known subunits (e.g., α1-6, β1-3, γ1-3), allows for the formation of numerous receptor subtypes with distinct anatomical distributions and physiological roles.[11] This heterogeneity provides the basis for subtype-selective pharmacology. Classical benzodiazepines, such as diazepam and lorazepam, are non-selective positive allosteric modulators (PAMs) that bind to the interface between an α and a γ subunit, enhancing the effect of GABA at receptors containing α1, α2, α3, or α5 subunits.[13] While highly effective as anxiolytics and anticonvulsants, their clinical utility is limited by a significant side-effect profile.[17] Decades of research using genetic knock-in animal models and selective compounds have successfully deconvoluted the functions of these α subunits [16]:
- α1 Subunit: Widely expressed and strongly linked to the sedative, amnesic, and ataxic effects of benzodiazepines.[13]
- α2 and α3 Subunits: Predominantly associated with the anxiolytic and anticonvulsant properties of these drugs.[13] The spinal cord expression of these subunits also implicates them in pain modulation.[13]
- α5 Subunit: Primarily located in the hippocampus and linked to cognitive processes, learning, and memory. Modulation of this subunit can lead to cognitive deficits.[13]
This functional segregation created the central hypothesis for a new generation of CNS drugs: a molecule that selectively modulates α2/α3-containing receptors while avoiding α1 and α5 could retain the therapeutic benefits of benzodiazepines while shedding their most problematic side effects. AZD-7325 was designed to be the clinical embodiment of this hypothesis.[9]
2.2 Mechanism of Action of AZD-7325
AZD-7325 is classified as a synthetic organic, partial positive allosteric modulator of the GABAA receptor.[3] Its mechanism is defined by both its high-affinity, selective binding and its specific functional activity at different receptor subtypes.
2.2.1 Subtype-Selective Binding Affinity
AZD-7325 exhibits a distinct binding profile characterized by high affinity for the therapeutically targeted subunits and significantly lower affinity for the α5 subunit, which is associated with cognitive side effects. Radioligand binding assays have quantified these affinities (Ki, inhibitor constant) as follows:
- GABAA α1: $K_i = 0.5$ nM [2]
- GABAA α2: $K_i = 0.3$ nM [2]
- GABAA α3: $K_i = 1.3$ nM [2]
- GABAA α5: $K_i = 230$ nM [2]
This profile demonstrates a clear preference for α1, α2, and α3 over α5. The selectivity ratio for the primary therapeutic target (α2) versus the primary cognitive liability target (α5) is over 750-fold ($230 \text{ nM} / 0.3 \text{ nM}$).
2.2.2 Subtype-Selective Functional Efficacy
Beyond simple binding, the functional consequence of that binding is what truly defines the drug's mechanism. Electrophysiological studies have shown that AZD-7325 has a unique functional signature:
- It acts as a partial agonist at GABAA receptors containing α2 and α3 subunits. This means it enhances the GABA response, but to a lesser degree than a full agonist like diazepam. Its efficacy is reported to be approximately 15-18% of the maximal response elicited by diazepam.[16]
- It acts as a neutral antagonist at the α1 subunit.[3] This means it binds to the α1-containing receptor but does not modulate its function, effectively blocking other benzodiazepine-site ligands without producing an effect itself. This property is key to its designed lack of sedation.
- It has minimal efficacy at the α5 subunit, consistent with its low binding affinity.[9]
This combined profile of high-affinity partial agonism at α2/α3 and neutral antagonism at α1 was intended to create a potent, non-sedating anxiolytic.[9]
Table 2: GABAA Receptor Subunit Binding Affinities (Ki) and Functional Efficacy of AZD-7325
| GABAA Subtype | Binding Affinity (Ki, nM) | Functional Activity (vs. Diazepam) | Source(s) |
|---|
| α1β3γ2 | 0.5 | Neutral Antagonist | 2 |
| α2β3γ2 | 0.3 | Partial Agonist (~15-18% max efficacy) | 2 |
| α3β3γ2 | 1.3 | Partial Agonist (~15-18% max efficacy) | 2 |
| α5β3γ2 | 230 | Low/Minimal Efficacy | 2 |
2.3 Pharmacokinetics, Metabolism, and Drug Interaction Profile
AZD-7325 was developed as an orally administered drug.[2] Nonclinical and early clinical studies characterized its absorption, distribution, metabolism, and excretion (ADME) properties, as well as its potential for drug-drug interactions (DDI).
2.3.1 Metabolism
The compound undergoes extensive metabolism in vivo. Studies across different species, facilitated by the synthesis of a carbon-14 labeled version ($[^{14}C]$AZD7325), identified a large number of metabolites.[2] These studies revealed complex metabolic pathways, including oxidative processes and a notable cyclization and aromatization pathway.[2]
2.3.2 Cytochrome P450 (CYP) Enzyme Interactions
The potential for AZD-7325 to interact with the CYP enzyme system, a major pathway for drug metabolism, was a critical area of investigation. The findings revealed a noteworthy discrepancy between in vitro predictions and in vivo clinical reality.
- In Vitro Studies: Initial screening using cultured human hepatocytes indicated that AZD-7325 was a moderate inducer of CYP1A2 mRNA and a potent inducer of CYP3A4 mRNA.[19] At concentrations of 1 to 10 µM, AZD-7325 produced CYP1A2 and CYP3A4 induction responses that were 17.9%-54.9% and 76.9%-85.7% of the positive control, respectively.[21] Such results would typically signal a high risk of clinically significant drug-drug interactions, potentially complicating co-administration with other medications.
- In Vivo Clinical Study: A subsequent clinical study was conducted in healthy volunteers to assess this risk directly.[21] Subjects received multiple 10 mg daily doses of AZD-7325 to reach steady-state, achieving a maximum plasma concentration ($C_{max}$) of approximately 0.2 µM (200 nM).[21] When co-administered with probe substrates, the results were markedly different from the in vitro predictions:
- CYP1A2 Activity: AZD-7325 had no effect on the pharmacokinetics of caffeine, a sensitive CYP1A2 substrate.[21]
- CYP3A4 Activity: AZD-7325 had only a weak inducing effect on CYP3A4 activity, decreasing the geometric mean area under the curve (AUC) of the sensitive substrate midazolam by just 19%.[21]
This divergence underscores a crucial principle in drug development: the clinical relevance of in vitro findings is critically dependent on the actual in vivo drug exposure. The concentrations used in the hepatocyte studies (1-10 µM) were 5- to 50-fold higher than the peak plasma concentrations achieved at a therapeutic dose (0.2 µM). Thus, while AZD-7325 possesses the intrinsic ability to induce these enzymes, it does not reach the necessary concentrations in the body to do so to a clinically meaningful extent. This in vivo study effectively de-risked a major potential liability for the compound.[21]
2.4 Preclinical Efficacy in Models of CNS Disorders
The unique pharmacological profile of AZD-7325 was tested in several preclinical animal models, which not only supported its initial development for anxiety but also provided the foundation for its later redevelopment.
- Anxiety: In early preclinical models, AZD-7325 demonstrated potent anxiolytic-like effects without the sedative side effects commonly seen with non-selective benzodiazepines.[2]
- Fragile X Syndrome (FXS): FXS is a neurodevelopmental disorder characterized by symptoms including sensory hypersensitivity and a high incidence of seizures, pointing to a state of neuronal hyperexcitability.[14] In the Fmr1 knockout (KO) mouse model of FXS, AZD-7325 (investigated under its new name, BAER-101) showed promising results. Treatment with the compound was found to:
- Reduce the hyperexcitability of cortical circuits.[14]
- Decrease susceptibility to audiogenic (sound-induced) seizures.[3]
- Partially correct abnormalities in cortical EEG power.[14]
- Improve performance in a novel object recognition memory task.14
These findings provided a strong biological rationale for investigating the drug in neurodevelopmental disorders associated with GABAergic dysfunction.
- Epilepsy: The most compelling preclinical efficacy data for AZD-7325/BAER-101 has emerged from models of epilepsy. In the Genetic Absence Epilepsy Rat from Strasbourg (GAERS) model, a well-validated model for absence seizures, oral administration of BAER-101 demonstrated profound and dose-dependent anti-seizure activity.[23]
- It achieved full suppression of seizure activity (spike-wave discharges) with a minimal effective dose (MED) of 0.3 mg/kg.[17]
- The effect was rapid in onset and sustained throughout the testing period.[23]
- Importantly, no adverse events were observed at doses up to 100 mg/kg, which is over 300 times the MED, suggesting a very wide therapeutic index for its anticonvulsant effects.17
This robust efficacy signal in a primary seizure model provides the cornerstone for the drug's current development strategy in epilepsy.
The pattern of these preclinical results is telling. While the compound showed anxiolytic-like activity, its most powerful and clear-cut effects were observed in conditions of overt neuronal hyperexcitability, such as seizure models. This suggests that the molecule's core biological strength lies in its ability to stabilize hyperactive neuronal networks, a property that aligns more directly with an anti-epileptic or neurostabilizing indication than with the more nuanced neurobiology of anxiety disorders.
2.5 Safety Pharmacology and Toxicology Summary
Nonclinical safety assessment is a critical component of drug development. Available information provides a partial view of AZD-7325's toxicology profile.
- GHS Hazard Classification: As a chemical entity supplied for research, AZD-7325 has an aggregated GHS classification from the European Chemicals Agency (ECHA) C&L Inventory. It is classified as Acute Toxicity, Oral (Category 4), carrying the hazard statement H302: "Harmful if swallowed," and Serious Eye Damage (Category 1), with H318: "Causes serious eye damage".[1] These classifications are standard for many chemical reagents and are primarily relevant to occupational handling rather than clinical use.
- Preclinical Toxicology: Detailed results from formal preclinical toxicology studies (e.g., repeat-dose toxicology in rodent and non-rodent species) are not available in the provided materials. However, the development history of the drug class provides some context. A similar subtype-selective GABAA PAM, TPA-023, was terminated during development due to toxicity observed in preclinical species, indicating a potential class-related risk that would have been carefully monitored for AZD-7325.[17] In contrast, the efficacy study in the GAERS rat model reported an excellent safety margin, with no adverse events observed at doses far exceeding the effective dose, which is a highly encouraging sign for its therapeutic index in an epilepsy context.[17]
Section 3: The Clinical Development of AZD-7325
The clinical journey of AZD-7325 is a story of two distinct phases: an initial, extensive program by AstraZeneca targeting anxiety, which ultimately failed to meet its primary goals, followed by a strategic revival for neurodevelopmental and seizure disorders. This section provides a comprehensive chronicle and analysis of the clinical trials that have defined the drug's trajectory.
3.1 Overview of the Clinical Program
AstraZeneca's initial investment in AZD-7325 was substantial, comprising at least 10 clinical studies that enrolled over 700 participants, including both healthy volunteers and patients with various CNS disorders.[5] The program progressed to Phase II, the highest stage of development reached under its original sponsor.[1] The trials spanned a range of objectives, from initial safety and dosing in Phase I to proof-of-concept efficacy studies in Phase II, as well as specialized studies to explore its CNS effects and potential in other indications.
3.2 Phase I Studies: Characterizing Safety, Tolerability, and CNS Effects in Healthy Volunteers
The Phase I program was designed to establish the foundational safety, tolerability, pharmacokinetic (PK), and pharmacodynamic (PD) profile of AZD-7325 in humans.
- Ascending Dose Studies: Standard single ascending dose (SAD) and multiple ascending dose (MAD) studies were conducted in healthy volunteers, including in specific populations like Japanese subjects, to determine the safe and tolerable dose range and to characterize the drug's PK profile.[26] These studies (e.g., NCT00681915, NCT00945425) were critical for selecting doses for subsequent Phase II trials.[26]
- CNS Pharmacodynamics and the "Anxio-Selective" Signature: A pivotal Phase I, randomized, double-blind, four-way crossover study (D1140C00003) provided the first clinical evidence supporting the drug's designed mechanism.[16] The study compared single oral doses of AZD7325 (2 mg and 10 mg) against placebo and the non-selective benzodiazepine lorazepam (2 mg) in 16 healthy males, using a battery of validated CNS tests.[16] The key findings were:
- Mitigated CNS Impairment: Lorazepam produced robust and statistically significant impairment across multiple domains, including cognitive, psychomotor, and neurophysiologic functions. In stark contrast, neither dose of AZD7325 induced statistically significant effects on these pharmacodynamic measurements.[16]
- Pharmacological Selectivity: To specifically test the "anxio-selective" hypothesis, researchers analyzed the relationship between saccadic peak velocity (SPV), an eye-movement metric considered a surrogate marker for anxiolytic GABAA activity, and measures of sedation like body sway and subjective alertness.[16] For lorazepam, reductions in SPV were linearly correlated with increased body sway and decreased alertness. For AZD7325, this relationship was significantly flatter, indicating that it could engage the target mechanism (affecting SPV) with a much lower penalty on sedation and postural stability.[16] This provided the first clinical validation of its differentiated, safer CNS profile compared to traditional benzodiazepines.
- Receptor Occupancy via PET Imaging: To confirm that the drug was reaching its target in the human brain, positron emission tomography (PET) studies were conducted using the radioligand $[^{11}C]$flumazenil, which binds to the benzodiazepine site on GABAA receptors.[20] These studies demonstrated a dose-dependent reduction in $[^{11}C]$flumazenil binding, confirming target engagement.[20] Crucially, high levels of receptor occupancy (over 80% at a 10 mg dose) were achieved without causing the sedation or cognitive impairment expected from a non-selective agent at similar occupancy levels.[20] This was a major success for the drug's design. However, an important observation was that even at these high occupancy levels, the measurable pharmacodynamic effects of AZD-7325 remained modest, leading to the conclusion that the drug might possess low intrinsic efficacy and that clinically effective concentrations could be higher than those predicted by receptor binding alone.[16]
3.3 The Pivotal Indication: Phase II Trials in Generalized Anxiety Disorder (GAD)
Based on the strong preclinical rationale and the promising Phase I data, AZD-7325 advanced into Phase II trials for GAD. This stage would prove to be the critical turning point in its development.
- Study Designs: At least two major proof-of-concept studies were conducted.
- D1140C00006 (NCT00808249): This was a large, multi-center, randomized, double-blind, placebo-controlled study that enrolled 424 patients with GAD. It tested three dose regimens of AZD7325 (2 mg BID, 5 mg BID, and 10 mg QD) against a placebo over a 28-day treatment period.[31]
- D1140C00014: This study enrolled 369 subjects and compared two doses of AZD7325 (5 mg BID and 15 mg BID) against both placebo and an active comparator, lorazepam (2 mg BID), over 28 days.[34]
- Primary Endpoint: The primary efficacy measure for these studies was the change from baseline to the end of treatment in the total score of the Hamilton Rating Scale for Anxiety (HAM-A), a standard clinical instrument for assessing anxiety severity.[33]
- Outcome and Analysis: The trials failed to meet their primary endpoint.[5] In the D1140C00006 study, while all treatment arms, including placebo, showed a numerical improvement (a reduction in HAM-A scores), the improvement in the AZD7325 groups was not statistically superior to that of the placebo group.[33] The failure to separate from a high placebo response is a common challenge in psychiatric drug trials. A post-hoc sub-analysis of the data, which excluded patients who dropped out or were non-compliant (as determined by plasma drug levels), suggested a potential dose-related anxiolytic signal.[5] However, such exploratory analyses cannot overcome the failure of the primary endpoint in the intent-to-treat population and were insufficient to support further development for GAD.
The failure in GAD, despite the successful engineering of a molecule with a superior safety profile, presents a critical paradox. The very selectivity that made AZD-7325 safer and more tolerable than older benzodiazepines may have simultaneously rendered it clinically ineffective as an anxiolytic. It is plausible that the broad CNS-dampening effects mediated by the α1 subunit, while responsible for sedation, are also an integral part of the therapeutic relief that patients with GAD experience with non-selective agents. The drug may have been too selective, a scientifically elegant molecule that failed to produce a robust enough biological effect to be perceived as efficacious in a complex clinical disorder with a high placebo response rate.
3.4 Exploratory Trials in Neurodevelopmental Disorders
In parallel with the main anxiety program, the unique mechanism of AZD-7325 prompted its investigation in neurodevelopmental disorders where GABAergic system dysfunction is implicated.
- Autism Spectrum Disorder (ASD): A Phase II proof-of-mechanism trial (NCT01966679) was initiated to evaluate AZD-7325 for social disability in young adults with ASD.[35] This study was a collaborative effort funded by the National Institute of Mental Health (NIMH) as part of its "Fast-Fail" initiative, designed to test novel mechanisms efficiently.[36] A key innovation of this trial was its use of quantitative electroencephalography (EEG) as a primary outcome measure to objectively assess target engagement in the CNS of the patient population, a strategy designed to provide a clear go/no-go signal for further development by avoiding the subjectivity and high placebo response of traditional behavioral scales.[36] This trial represented a forward-thinking approach to de-risking CNS drug development.
- Fragile X Syndrome (FXS): Building on the strong preclinical data, a Phase I/II, randomized, double-blind, crossover study (NCT03140813) was conducted to investigate the safety, tolerability, and efficacy of AZD7325 (at 5 mg and 15 mg BID doses) in adults with FXS.[39] This study aimed to translate the promising findings from the Fmr1 KO mouse model into a clinical setting.[40]
3.5 Specialized Human Studies
Two additional specialized studies further characterized the unique profile of AZD-7325.
- Human Abuse Liability: To formally assess its potential for abuse, a dedicated Phase I study (NCT00902772) was conducted in healthy recreational users of CNS depressants.[41] The results of this study supported the hypothesis that AZD7325 has a lower abuse potential than lorazepam, a key differentiating feature from traditional benzodiazepines.[5]
- Cutaneous Sensation and Neuropathic Pain: A Phase I biomarker study (Eudract: 2015-000642-35) was designed to explore a novel hypothesis: whether enhancing central GABAergic inhibition with a non-sedating drug like AZD7325 could improve sensory and motor function.[25] The study in healthy volunteers investigated effects on manual dexterity and skin sensation, with the long-term rationale that if successful, it could provide a basis for treating conditions with peripheral sensory deficits, such as peripheral neuropathies, for which there are limited symptomatic treatments.[25]
A summary of the major clinical trials conducted for AZD-7325 is provided in Table 3.
Table 3: Summary of Major Clinical Trials for AZD-7325 / BAER-101
| Trial ID | Phase | Indication | Subjects (N) | Doses Studied | Primary Endpoint | Outcome/Status | Source(s) |
|---|
| D1140C00003 | 1 | Healthy Volunteers | 16 | 2 mg, 10 mg (single dose) | Safety, CNS Pharmacodynamics | Favorable safety profile; demonstrated "anxio-selective" signature vs. lorazepam | 16 |
| NCT00681915 | 1 | Healthy Volunteers | N/A | Multiple Ascending Doses | Safety and Tolerability | Completed | 26 |
| NCT00902772 | 1 | Recreational CNS Depressant Users | N/A | N/A | Abuse Potential | Completed; lower abuse potential than lorazepam | 5 |
| PET Study | 1 | Healthy Volunteers | 12 | 0.2 mg to 30 mg | GABAA Receptor Occupancy | High receptor occupancy achieved without sedation | 20 |
| D1140C00006 (NCT00808249) | 2 | Generalized Anxiety Disorder (GAD) | 424 | 2 mg BID, 5 mg BID, 10 mg QD | Change in HAM-A Score | Completed; Failed to meet primary endpoint vs. placebo | 31 |
| D1140C00014 | 2 | Generalized Anxiety Disorder (GAD) | 369 | 5 mg BID, 15 mg BID | Change in HAM-A Score | Completed; Failed to meet primary endpoint | 5 |
| NCT01966679 | 2 | Autism Spectrum Disorder (ASD) | N/A | N/A | EEG Biomarkers | Completed | 35 |
| NCT03140813 | 1/2 | Fragile X Syndrome (FXS) | N/A | 5 mg BID, 15 mg BID | Safety, Tolerability, Efficacy Measures | Completed | 39 |
| EudraCT: 2015-000642-35 | 1 | Healthy Volunteers | N/A | Single Dose | Effect on Cutaneous Sensation | Completed | 25 |
Section 4: Integrated Clinical Safety and Tolerability Analysis
A thorough understanding of a drug's safety profile is paramount. The extensive clinical program for AZD-7325, involving over 700 individuals, provides a robust dataset from which to evaluate its safety and tolerability, both in absolute terms and relative to the existing standard of care.
4.1 Summary of Adverse Events Across the Clinical Program
Across all studies, AZD-7325 was generally reported to be safe and well-tolerated.[5] The adverse event (AE) profile was consistent and predictable based on its mechanism of action as a GABAergic modulator.
- Most Common Adverse Events: The most frequently reported side effects across multiple studies, including those in patients with GAD and ASD, were mild to moderate in severity and included dizziness, sleepiness (somnolence), fatigue, and headache.[5] In some early studies, subjects also reported feeling "hot," being in an "especially happy mood," or "feeling drunk".[38] These are all characteristic effects of CNS depressants.
4.2 Serious Adverse Events and Trial Discontinuations
While common AEs were manageable, the rate of discontinuation due to AEs provides insight into the drug's dose-limiting toxicities.
- Serious Adverse Events (SAEs): SAEs were infrequent. In the large 424-patient GAD study (D1140C00006), only four subjects (0.9%) experienced an SAE, and notably, none of these occurred in the high-dose (10 mg QD) or placebo groups.[33] No deaths were reported in any of the clinical trials.[33]
- Discontinuations due to Adverse Events: The primary reason for subjects withdrawing prematurely from clinical trials was the experience of an adverse event.[33] In the D1140C00006 study, 16.5% of subjects withdrew early, with AEs accounting for 38.6% of these discontinuations.[33] The most common AEs leading to withdrawal were dizziness and mental impairment, followed by sedation and somnolence.[33] A slightly higher number of subjects discontinued due to an AE in the 10 mg QD group compared to the lower dose and placebo groups, establishing these on-target CNS effects as the primary dose-limiting factors.[33]
The safety profile reveals a well-defined and predictable therapeutic window. The dose-limiting side effects are not idiosyncratic or related to off-target toxicity but are rather an extension of the drug's primary GABAergic mechanism of action. While AZD-7325 is significantly better tolerated than non-selective benzodiazepines, it is not devoid of CNS-depressant effects at higher doses. This establishes a clear ceiling on tolerability that must be respected in the design of future clinical trials. The central challenge for its redevelopment is to identify an indication where the dose required for robust efficacy falls comfortably below the doses that lead to these limiting, yet predictable, side effects.
4.3 Comparative Safety: The Anxio-Selective Advantage Over Benzodiazepines
A key objective of the AZD-7325 program was to demonstrate a superior safety profile compared to non-selective benzodiazepines. The clinical data strongly support the success of this aspect of its design.
- Reduced CNS Impairment: Direct head-to-head comparisons in Phase I studies conclusively showed that AZD-7325 produced a significantly mitigated side-effect burden compared to lorazepam.[16] At doses that achieved high GABAA receptor occupancy in the brain, AZD-7325 did not cause the profound cognitive, psychomotor, and sedative impairments observed with lorazepam.[16]
- Lower Abuse Potential: A dedicated human abuse liability study (NCT00902772) in a population of recreational CNS depressant users provided formal evidence that the risk of abuse with AZD-7325 was lower than that of lorazepam.[5] This is a critical differentiating factor, as the abuse and dependence potential of benzodiazepines severely restricts their long-term use.
- Validation of the Design Hypothesis: The PET imaging studies provided the most direct validation of the safety hypothesis.[20] The ability to achieve high (>80%) target engagement in the brain without inducing significant sedation was a landmark finding, confirming that the selective pharmacological profile designed in vitro had successfully translated into a differentiated and safer clinical profile in humans.[20]
Section 5: Regulatory, Commercial, and Intellectual Property History
The trajectory of AZD-7325 from a promising asset within a major pharmaceutical company to a repurposed candidate in a smaller biotech provides a compelling look into the strategic and economic realities of drug development. This section details its corporate history, the rationale for its initial discontinuation, its subsequent revival, and the intellectual property landscape that underpins its value.
5.1 Discovery and Early Development at AstraZeneca
AZD-7325 emerged from AstraZeneca's CNS research programs in the mid-2000s, part of a broader industry effort to develop safer, next-generation GABAA receptor modulators.[18] The medicinal chemistry effort focused on the cinnoline scaffold, optimizing it for the desired subtype-selective profile.[18] The first patent applications covering the compound were filed around 2007, and the clinical development program was initiated shortly thereafter, with the first Phase I studies beginning in 2008.[1]
5.2 Discontinuation Post-GAD and the Rationale
Following the completion of the Phase II proof-of-concept studies in Generalized Anxiety Disorder around 2009-2010, the development of AZD-7325 for this indication was halted.[33] While AstraZeneca has not issued a formal public statement detailing the specific reasons for the discontinuation, the clinical trial results provide a clear rationale. The failure of the drug to meet its primary efficacy endpoints in two large, well-controlled GAD trials made it an unviable candidate for a large-market indication like anxiety.[5] In the high-risk, high-reward environment of pharmaceutical R&D, a lack of efficacy in a pivotal trial is a standard and definitive reason for terminating a program, regardless of a favorable safety profile.[17] The asset was subsequently shelved.
5.3 The Second Act: Re-emergence as BAER-101
The story of AZD-7325 exemplifies a modern, efficient model of value creation in the biopharmaceutical ecosystem, where promising but deprioritized assets from large companies find new life in smaller, more focused entities.
In December 2019, Fortress Biotech announced that its newly formed partner company, Baergic Bio, Inc., had executed an exclusive worldwide licensing agreement with AstraZeneca to acquire the rights to AZD7325.[6] This transaction was characteristic of an "asset-centric" biotech strategy:
- Asset Acquisition: Baergic Bio was formed with the primary purpose of developing this specific asset.[6]
- Terms of the Deal: In exchange for the rights, AstraZeneca received an equity position in the new company, aligning its interests with the future success of the compound. Crucially, the deal included the complete preclinical and clinical data package, which represented hundreds of millions of dollars in prior R&D investment and contained invaluable information on the drug's safety, PK, and biological activity.[6]
- Renaming and Repositioning: The compound was renamed BAER-101 to mark its new chapter of development.[6]
- Academic Collaboration: Baergic Bio also concurrently entered into an agreement with Cincinnati Children's Hospital Medical Center, a leader in pediatric neuroscience research, to help advance the clinical development of BAER-101, likely leveraging the hospital's expertise and the promising data generated in Fragile X Syndrome.[6]
This strategic move allowed a well-characterized, de-risked molecule to be repurposed for indications where its biological profile was a better fit, a model that is far more capital-efficient than discovering a new chemical entity from scratch.
5.4 Current Development Strategy and Target Indications
The development of BAER-101 is now being advanced by Avenue Therapeutics. The strategic focus has pivoted completely away from anxiety, which is now listed as an inactive indication.[17] The current development plan is squarely focused on CNS disorders characterized by neuronal hyperexcitability, primarily epilepsy and related seizure disorders.[17] This strategy is directly supported by the highly compelling preclinical efficacy data in the GAERS rat model of absence seizures and the anti-seizure activity observed in the mouse model of Fragile X Syndrome.[14]
5.5 Patent Landscape and Market Exclusivity
A robust intellectual property (IP) portfolio is essential for securing the commercial potential of a drug candidate. The IP landscape for AZD-7325/BAER-101 includes patents covering its composition of matter, formulations, and methods of use.
- Composition of Matter: The original patents filed by AstraZeneca, such as EP1966158B1, claim the chemical structure of 4-amino-8-(2-fluoro-6-methoxyphenyl)-N-propylcinnoline-3-carboxamide and its use in treating CNS disorders, including anxiety.[43] These foundational patents provide the core market exclusivity for the molecule itself.
- Formulations: Subsequent patent applications were filed to protect specific pharmaceutical formulations of the drug, including solid oral dosage forms (US Patent Application 20090215744) and liquid formulations (US Patent Application 20090069292).[44]
- Methods of Use: As new potential uses for the drug were identified, new method-of-use patents have been filed. A key example is patent application US20150313913A1, filed by the University of Washington, which describes the use of α2/α3 selective GABAA PAMs for treating Autism Spectrum Disorders.[45] This type of IP is critical for protecting the commercial viability of the drug in its new, repurposed indications. The portfolio also includes patents covering combinations with other drugs.[46]
Section 6: Expert Analysis and Future Outlook
The comprehensive history of AZD-7325/BAER-101 offers a rich dataset for critical analysis, providing valuable lessons for CNS drug development and informing the future trajectory of the compound. This final section synthesizes the preceding information to offer a retrospective on its initial failure, an evaluation of its current scientific rationale, and an outlook on the challenges and opportunities that lie ahead.
6.1 A Critical Retrospective: Why Did AZD-7325 Fail in Anxiety?
The failure of AZD-7325 to demonstrate efficacy in Generalized Anxiety Disorder, despite its elegant design and successful target engagement, was likely the result of a confluence of pharmacological and clinical factors.
- The "Anxio-Selective" Hypothesis Paradox: The program's foundational hypothesis—that pure α2/α3 modulation could replicate the anxiolytic effects of benzodiazepines without α1-mediated sedation—was a scientific success from a safety perspective but a clinical failure from an efficacy perspective. The data suggest that the subtle, broad CNS-dampening effects provided by α1 agonism, while causing sedation, may be inseparable from the overall therapeutic effect perceived by patients as anxiety relief. The drug's "clean" profile may have been too clean to produce a robust clinical signal in GAD.
- Low Intrinsic Efficacy: The observation from PET studies that high receptor occupancy yielded only modest pharmacodynamic effects points to the possibility of low intrinsic efficacy.[16] As a partial agonist, AZD-7325 may not have been capable of amplifying the GABAergic signal sufficiently to overcome the pathological circuit dysfunction in GAD, even when bound to the correct targets.
- The Challenge of GAD Trials: Clinical trials in GAD are notoriously difficult due to high placebo response rates and the subjective nature of the primary endpoints (e.g., HAM-A scale).[47] A drug with a subtle effect, like AZD-7325, would have a very difficult time demonstrating statistical superiority over a strong placebo response, as was observed in the Phase II studies.[33]
6.2 The Scientific Rationale for Redevelopment in Epilepsy and Other CNS Disorders
In contrast to the ambiguity of its anxiolytic potential, the scientific rationale for developing BAER-101 as an anticonvulsant is exceptionally strong and data-driven.
- Direct Mechanism-Pathophysiology Match: Epilepsy is fundamentally a disorder of neuronal hyperexcitability. The primary mechanism of BAER-101—enhancing GABAergic inhibition—directly counteracts this core pathophysiology. This represents a much more direct and less speculative link between mechanism and disease than its application in anxiety.
- Compelling and Translatable Preclinical Data: The preclinical results in seizure models are not merely "promising"; they are definitive. The complete suppression of spike-wave discharges in the GAERS model at doses with a >300-fold safety margin is a powerful demonstration of efficacy in a highly validated and translatable model of epilepsy.[17] Similarly, the reduction of seizure susceptibility in the FXS mouse model further strengthens its potential in neurodevelopmental disorders associated with epilepsy.[14]
- Clinical Validation of Target Engagement: The extensive Phase I data has already established that the drug enters the human brain, engages its target receptor, and does so with a favorable safety profile.[20] This significantly de-risks the early stages of its redevelopment for epilepsy.
6.3 Future Challenges and Opportunities for BAER-101
The path forward for BAER-101 is promising but not without significant hurdles.
- Challenges:
- Clinical Efficacy and Dosing: The primary challenge will be to demonstrate robust anti-seizure efficacy in human epilepsy trials. This will require careful dose-finding studies to identify a dose that is effective without exceeding the tolerability ceiling defined by its on-target CNS side effects (dizziness, somnolence). The question of whether its partial agonism is sufficient for seizure control in humans remains to be answered.
- Competitive Landscape: The market for anti-epileptic drugs is mature and competitive. BAER-101 will need to demonstrate a clear advantage, likely through a superior safety and tolerability profile compared to existing GABAergic agents and other anti-seizure medications.
- Development Costs and Funding: As with any clinical-stage biotech asset, securing the substantial funding required to conduct pivotal Phase II and III trials is a constant challenge.[17]
- Opportunities:
- Best-in-Class Safety Profile: If the favorable safety profile holds at efficacious anti-seizure doses, BAER-101 has the potential to become a best-in-class GABAergic therapy, offering an alternative for patients who cannot tolerate the side effects of older drugs.
- Rare Disease Indications: Pursuing indications like Dravet Syndrome or Fragile X Syndrome could provide access to orphan drug incentives, smaller and faster clinical trials, and a more streamlined regulatory path to market.[22]
- Leveraging the Existing Data Package: The greatest opportunity lies in the immense value of the existing data from AstraZeneca. The safety database from over 700 subjects dramatically accelerates the development timeline and reduces risk, allowing the new sponsors to proceed directly to proof-of-concept studies in new indications without repeating extensive Phase I work.
6.4 Concluding Remarks
AZD-7325, now reborn as BAER-101, is more than just an investigational drug; it is a powerful illustration of the evolution of pharmaceutical science and strategy. Its story encapsulates the journey from a hypothesis-driven design based on molecular neuroscience to the pragmatic realities of clinical efficacy, and finally to a data-driven revival guided by a deeper understanding of the molecule's true strengths. It serves as a crucial reminder that in drug development, a clinical "failure" is not always an indictment of the molecule itself, but often an indication of a mismatch between the drug and the chosen disease. The future success of BAER-101 will ultimately depend on the ability of its new stewards to prove that its elegant pharmacology and compelling preclinical profile can finally translate into meaningful clinical benefit for patients with epilepsy and other disorders of neuronal hyperexcitability.
Works cited
- Azd-7325 | C19H19FN4O2 | CID 23581869 - PubChem - NIH, accessed October 24, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Azd7325
- Synthesis of C‐14 labeled GABAA α2/α3 selective partial agonists and the investigation of late‐occurring and long‐circulating metabolites of GABAA receptor modulator AZD7325 - PMC - PubMed Central, accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5969218/
- AZD 7325 (BAER-101, CAS Number: 942437-37-8) | Cayman ..., accessed October 24, 2025, https://www.caymanchem.com/product/36449/azd-7325
- AZD7325 | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed October 24, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=7712
- P117119_Avenue Therapeutics, accessed October 24, 2025, https://avenuetx.com/wp-content/uploads/2023/01/ATXI-Corporate-Presentation-January-2023.pdf
- Fortress Biotech Announces Exclusive Worldwide License ..., accessed October 24, 2025, https://www.fortressbiotech.com/news-media/press-releases/detail/243/fortress-biotech-announces-exclusive-worldwide-license
- List of investigational anxiety disorder drugs - Wikipedia, accessed October 24, 2025, https://en.wikipedia.org/wiki/List_of_investigational_anxiety_disorder_drugs
- AZD7325 | GABA Receptor modulator | Mechanism | Concentration - Selleck Chemicals, accessed October 24, 2025, https://www.selleckchem.com/products/azd-7325.html
- AZD7325 | GABAA α2/α3 PAM | Probechem Biochemicals, accessed October 24, 2025, http://www.probechem.com/products_AZD7325.html
- AZD-7325 (AZD7325) | CAS 942437-37-8 | AbMole BioScience ..., accessed October 24, 2025, https://www.abmole.com/products/azd-7325.html
- Subtype selective y-Aminobutyric Acid Type A Receptor.pdf - -ORCA - Cardiff University, accessed October 24, 2025, https://orca.cardiff.ac.uk/id/eprint/128150/1/Subtype%20selective%20y-Aminobutyric%20Acid%20Type%20A%20Receptor.pdf
- GABAkines – Advances in the Discovery, Development, and Commercialization of Positive Allosteric Modulators of GABAA Receptors - PubMed Central, accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9787737/
- Design and Identification of a Novel, Functionally Subtype Selective GABAA Positive Allosteric Modulator (PF-06372865) | Journal of Medicinal Chemistry - ACS Publications, accessed October 24, 2025, https://pubs.acs.org/doi/10.1021/acs.jmedchem.9b00322
- GABAA Alpha 2,3 Modulation Improves Select Phenotypes in a Mouse Model of Fragile X Syndrome - Frontiers, accessed October 24, 2025, https://www.frontiersin.org/journals/psychiatry/articles/10.3389/fpsyt.2021.678090/full
- AZD-7325: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 24, 2025, https://go.drugbank.com/drugs/DB13994
- The central nervous system effects of the partial GABA-Aα2,3-selective receptor modulator AZD7325 in comparison with lorazepam in healthy males, accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4256620/
- AZD-7325 - Drug Targets, Indications, Patents ... - Patsnap Synapse, accessed October 24, 2025, https://synapse.patsnap.com/drug/6c9f539cfa81436abddedb65913decea
- Subtype Selective γ-Aminobutyric Acid Type A Receptor (GABA A R) Modulators Acting at the Benzodiazepine Binding Site - SciSpace, accessed October 24, 2025, https://scispace.com/pdf/subtype-selective-g-aminobutyric-acid-type-a-receptor-gaba-a-1uyq8q12mn.pdf
- AZD7325 | GABAA Receptor Modulator - MedchemExpress.com, accessed October 24, 2025, https://www.medchemexpress.com/azd7325.html
- GABAA receptor occupancy by subtype selective GABAAα2,3 modulators: PET studies in humans - PubMed Central, accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5263201/
- A clinical study to assess CYP1A2 and CYP3A4 induction by AZD7325, a selective GABA(A) receptor modulator - an in vitro and in vivo comparison - PubMed, accessed October 24, 2025, https://pubmed.ncbi.nlm.nih.gov/22122233/
- GABAA Alpha 2,3 Modulation Improves Select Phenotypes in a ..., accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8175776/
- BAER-101 / AstraZeneca, Fortress - LARVOL DELTA, accessed October 24, 2025, https://delta.larvol.com/Products/?ProductId=7df3174e-0324-4964-84ee-f4ec577fcd06
- The Central Nervous System Effects Of the Partial GABA-Aα2,3 -Selective Receptor Modulator AZD7325 In Comparison With Lorazepam In Healthy Males. | Request PDF - ResearchGate, accessed October 24, 2025, https://www.researchgate.net/publication/262108131_The_Central_Nervous_System_Effects_Of_the_Partial_GABA-Aa23_-Selective_Receptor_Modulator_AZD7325_In_Comparison_With_Lorazepam_In_Healthy_Males
- Effects of AZD7325 on cutaneous sensation - Health Research ..., accessed October 24, 2025, https://www.hra.nhs.uk/planning-and-improving-research/application-summaries/research-summaries/effects-of-azd7325-on-cutaneous-sensation/
- Study Details | NCT00681915 | AZD7325 Multiple Ascending Dose ..., accessed October 24, 2025, https://clinicaltrials.gov/study/NCT00681915
- AZD-7325 Completed Phase 1 Trials for Healthy Volunteers (HV) Treatment - DrugBank, accessed October 24, 2025, https://go.drugbank.com/drugs/DB13994/clinical_trials?conditions=DBCOND0114743&phase=1&purpose=treatment&status=completed
- Study Details | NCT01906385 | Maximum Tolerated Dose, Safety ..., accessed October 24, 2025, https://clinicaltrials.gov/study/NCT00945425
- The central nervous system effects of the partial GABA-Aalpha2,3 -selective receptor modulator AZD7325 in comparison with lorazepam in healthy males. - DrugBank, accessed October 24, 2025, https://go.drugbank.com/articles/A33292
- study synopsis, accessed October 24, 2025, https://filehosting.pharmacm.com/DownloadService.ashx?client=CTR_MED_6111&studyid=1728&filename=CSR-D1140C00003.pdf
- AZD7325 Proof of Concept in Patients With Generalized Anxiety Disorder (GAD), accessed October 24, 2025, https://ctv.veeva.com/study/azd7325-proof-of-concept-in-patients-with-generalized-anxiety-disorder-gad
- AZD7325 Proof of Concept in Patients with Generalized Anxiety Disorder (GAD), accessed October 24, 2025, https://www.astrazenecaclinicaltrials.com/study/D1140C00006/
- d1140c00006 study synopsis, accessed October 24, 2025, https://filehosting.pharmacm.com/DownloadService.ashx?client=CTR_MED_6111&studyid=250&filename=CSR-D1140C00006.pdf
- Proof of Concept in Patients with Generalized Anxiety Disorder (GAD)AZD7325, accessed October 24, 2025, https://www.astrazenecaclinicaltrials.com/study/D1140C00014/
- AZD-7325 Completed Phase 2 Trials for Autism Spectrum Disorder ..., accessed October 24, 2025, https://go.drugbank.com/drugs/DB13994/clinical_trials?conditions=DBCOND0006160&phase=2&purpose=treatment&status=completed
- De-risking Psychiatric Drug Development: The NIMH's Fast Fail ..., accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5007185/
- Effects of GABA Modulator AZD7325 on Cutaneous Sensation | Cochrane Library, accessed October 24, 2025, https://www.cochranelibrary.com/content?templateType=related&urlTitle=/central/doi/10.1002/central/CN-01580389&doi=10.1002/central/CN-01580389&p_p_id=scolariscontentdisplay_WAR_scolariscontentdisplay&_scolariscontentdisplay_WAR_scolariscontentdisplay_action=related-content&p_p_lifecycle=0&p_p_mode=view&type=central&contentLanguage=
- Medication Study for ASD - Marcus Autism Center, accessed October 24, 2025, https://www.marcus.org/research/~/media/Marcus/Documents/Research/Medication-Study-For-ASD.pdf
- An Initial Study of AZD7325 in Adults With Fragile X Syndrome | ClinicalTrials.gov, accessed October 24, 2025, https://clinicaltrials.gov/study/NCT03140813
- AZD-7325 Completed Phase 1 Trials for Fragile X Syndrome Treatment | DrugBank Online, accessed October 24, 2025, https://go.drugbank.com/drugs/DB13994/clinical_trials?conditions=DBCOND0002392&phase=1&purpose=treatment&status=completed
- Lorazepam Completed Phase 1 Trials for Recreational CNS ..., accessed October 24, 2025, https://go.drugbank.com/drugs/DB00186/clinical_trials?conditions=DBCOND0043791&phase=1&status=completed
- AstraZeneca quietly sweeps out some notable mid-stage drug ..., accessed October 24, 2025, https://www.fiercebiotech.com/r-d/astrazeneca-quietly-sweeps-out-some-notable-mid-stage-drug-programs
- SUBSTITUTED CINNOLINE DERIVATIVES AS ... - Googleapis.com, accessed October 24, 2025, https://patentimages.storage.googleapis.com/41/e8/82/9d95928d516e6c/EP1966158B1.pdf
- Patents | Creekmore Pharma Consulting, accessed October 24, 2025, https://creekmorepharma.com/patents
- US20150313913A1 - Positive allosteric modulators of the gaba-a ..., accessed October 24, 2025, https://patents.google.com/patent/US20150313913A1/en
- FWP - 10/06/2022 - Avenue Therapeutics, Inc., accessed October 24, 2025, https://ir.avenuetx.com/sec-filings/all-sec-filings/content/0001104659-22-106772/0001104659-22-106772.pdf
- Pharmacotherapy of Anxiety Disorders: Current and Emerging Treatment Options | Focus, accessed October 24, 2025, https://psychiatryonline.org/doi/10.1176/appi.focus.19203