MedPath

Colestilan chloride Advanced Drug Monograph

Published:Oct 12, 2025

Drug Type

Small Molecule

CAS Number

95522-45-5

Associated Conditions

Hyperphosphataemia

Nelonicline (ABT-126): A Comprehensive Pharmacological and Clinical Development Review of a Selective α7 Nicotinic Acetylcholine Receptor Agonist

I. Executive Summary

Nelonicline, also known by its developmental code ABT-126, is an investigational small molecule drug originated by Abbott Laboratories (later AbbVie). It was designed as an orally active, selective agonist of the alpha-7 nicotinic acetylcholine receptor (α7 nAChR), a key modulator of neuronal signaling in the central nervous system. The development of nelonicline was founded on the well-established cholinergic hypothesis, which links cognitive deficits in neurodegenerative and psychiatric disorders, such as Alzheimer's disease (AD) and schizophrenia, to dysfunction in the brain's acetylcholine systems. By selectively targeting the α7 nAChR, nelonicline was intended to enhance cholinergic transmission, thereby improving cognitive functions like memory, learning, and attention.

The compound demonstrated a promising preclinical and early clinical profile, characterized by high-affinity binding to its target, desirable pharmacokinetic properties including oral activity and brain permeability, and a metabolic pathway that minimized the potential for common drug-drug interactions. Across a broad Phase 2 clinical program, nelonicline consistently exhibited a favorable safety and tolerability profile, with adverse events generally being mild to moderate and comparable in frequency to placebo.

Despite these advantageous characteristics, nelonicline ultimately failed to demonstrate clinical efficacy. In multiple large, well-controlled Phase 2 trials for mild-to-moderate AD, the drug did not achieve statistically significant improvement on the primary cognitive endpoint, the Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog), when used as either a monotherapy or as an adjunctive treatment to standard-of-care acetylcholinesterase inhibitors. Similarly, in extensive Phase 2b studies targeting cognitive impairment associated with schizophrenia (CIAS), nelonicline failed to show a pro-cognitive effect on the MATRICS Consensus Cognitive Battery (MCCB) in both smoking and non-smoking patient populations. An intriguing but statistically non-significant trend toward improvement in negative symptoms was observed in the schizophrenia trials, but this was insufficient to support further development.

Consequently, due to a consistent and definitive lack of efficacy across both of its primary indications, the clinical development of nelonicline was discontinued. Its trajectory serves as a significant case study in CNS drug development, illustrating the profound challenge of translating a sound pharmacological rationale and promising preclinical data into tangible therapeutic benefits for patients with complex neuropsychiatric disorders.

II. Chemical Identity and Pharmaceutical Classification

Nelonicline is a new molecular entity classified as a small molecule drug.[1] Its chemical identity is defined by a unique combination of structural motifs designed for central nervous system activity.

Nomenclature and Identifiers

  • Generic Name: Nelonicline [3]
  • Developmental Code: ABT-126 [2]
  • Systematic (IUPAC) Name: 2-(1-azatricyclo[3.3.1.13,7]decan-4-yloxy)-5-phenyl-1,3,4-thiadiazole [3]
  • CAS Registry Numbers: Multiple numbers are associated with the compound, including 1026687-03-5 and 1026134-63-3, which may correspond to different salt forms or stereoisomers studied during development.[3]

Chemical Formula and Molecular Weight

  • Molecular Formula: .[3] This formula is the most consistently reported in authoritative chemical databases.
  • Molecular Weight: Approximately 313.4 g/mol.[3]

Structural and Drug Classifications

Nelonicline's structure is a composite of a rigid cage-like system and aromatic heterocyclic rings. Chemically, it is classified as an Adamantane, an Aza compound, and a Thiadiazole.[1] The presence of the adamantane moiety is a deliberate and significant feature of its molecular architecture. Adamantane is a classic pharmacophore frequently incorporated into CNS drug candidates to increase lipophilicity and structural rigidity. This bulky, three-dimensional hydrocarbon cage enhances a molecule's ability to passively diffuse across the lipid-rich blood-brain barrier, a critical requirement for drugs targeting receptors within the brain. Its inclusion in nelonicline's structure reflects a rational design approach aimed at ensuring sufficient CNS exposure to engage its target receptor.

From a therapeutic perspective, nelonicline is classified as a Nootropic, or cognition-enhancing, agent, reflecting its intended clinical application.[1]

III. Pharmacological Profile: Targeting the α7 Nicotinic Receptor

The therapeutic hypothesis for nelonicline was based on its specific interaction with the α7 subtype of the nicotinic acetylcholine receptor, a key component of the cholinergic system implicated in cognitive function and neuroinflammation.

Mechanism of Action

Nelonicline is a selective alpha-7 nicotinic acetylcholine receptor (α7 nAChR) agonist.[1] Some reports further refine this classification to a partial agonist or allosteric modulator, which more accurately reflects its functional activity.[7] It exerts its effect by binding to and activating the CHRNA7 protein, a ligand-gated ion channel, thereby mimicking the action of the endogenous neurotransmitter acetylcholine.[2]

Receptor Binding Affinity and Potency

Nelonicline demonstrates potent engagement with its primary target. In vitro studies have shown that it binds with high affinity to α7 nAChRs in human brain tissue, with a reported inhibition constant () of 12.3 nM.[4]

Functional assays confirm its activity as a partial agonist. In Xenopus oocytes engineered to express human α7 nAChRs, nelonicline was shown to activate receptor currents with a half-maximal effective concentration () of 2 µM. Its maximal effect was measured at 74% of the response elicited by acetylcholine, defining it as a partial agonist because its intrinsic activity is less than that of the full endogenous agonist.[4]

A notable feature of nelonicline's pharmacological profile is the significant difference between its high binding affinity ( = 12.3 nM) and its substantially lower functional potency ( = 2000 nM). This discrepancy of over 160-fold suggests a relatively inefficient coupling between receptor occupancy and channel activation. While the drug can effectively bind to the receptor at low nanomolar concentrations, a much higher concentration is required to induce a robust functional response. This characteristic may have been a contributing factor to its lack of efficacy in clinical settings, as therapeutically achievable brain concentrations, while sufficient for receptor binding, may have been insufficient to produce the degree of receptor activation needed for a pro-cognitive effect.

Receptor Selectivity Profile

Nelonicline exhibits a favorable selectivity profile for the α7 nAChR over other related receptors. Although it does bind to the α3β4* nAChR subtype with a  of 60 nM, its functional activity at this receptor is negligible, showing only 12% efficacy at an extremely high concentration of 100,000 nM.[4] The drug also functions as an antagonist at the serotonin 5-HT3 receptor, a property shared by some other α7 nAChR modulators. However, its affinity for the 5-HT3 receptor ( = 140 nM) is more than tenfold lower than its affinity for the α7 nAChR, indicating that its primary action at therapeutic concentrations would be mediated through the α7 receptor.[4]

Scientific Rationale for Therapeutic Targeting

The development of nelonicline was firmly grounded in the cholinergic hypothesis of cognitive dysfunction, which proposes that a decline in acetylcholine signaling is a key contributor to the cognitive and memory deficits seen in Alzheimer's disease and schizophrenia.[10] Nicotinic acetylcholine receptors, particularly the α7 subtype, are densely expressed in brain regions essential for higher-order cognitive processes, including the hippocampus and cerebral cortex.[10]

The α7 nAChR is a homopentameric (composed of five identical α7 subunits) ligand-gated ion channel with unique properties, including high permeability to calcium ions () and a characteristic rapid desensitization following activation.[6] Activation of these receptors is known to modulate the release of other key neurotransmitters, such as glutamate, GABA, and dopamine, and to play a critical role in synaptic plasticity, the cellular basis of learning and memory.[6] Furthermore, the α7 nAChR is a crucial component of the cholinergic anti-inflammatory pathway, a neuro-immune signaling axis that regulates inflammation.[6] The therapeutic rationale was therefore twofold: direct activation of α7 nAChRs was hypothesized to enhance cognitive processing, while also potentially mitigating the neuroinflammatory processes that contribute to the pathology of AD.

However, the inherent rapid desensitization of the α7 nAChR presents a significant challenge for therapeutic strategies based on direct agonism. Continuous stimulation by an agonist like nelonicline can drive the receptor into a prolonged, non-functional (desensitized) state, potentially negating or even reversing any therapeutic benefit. This fundamental property of the target receptor may explain the failure of nelonicline and other similar agonists. It also provides the context for the strategic shift in the field toward developing positive allosteric modulators (PAMs), which do not directly activate the receptor but rather enhance its response to endogenous acetylcholine, a mechanism that may be less prone to inducing profound desensitization.[2]

Table 1: Key Physicochemical and Pharmacological Properties of Nelonicline

ParameterValue / DescriptionSource(s)
Generic NameNelonicline3
Developmental CodeABT-1262
Molecular Formula3
Molecular Weight~313.4 g/mol3
Chemical ClassAdamantane; Aza compound; Thiadiazole1
Pharmaceutical ClassNootropic; Small Molecule1
Primary TargetAlpha-7 Nicotinic Acetylcholine Receptor (α7 nAChR; CHRNA7)1
Mechanism of ActionSelective α7 nAChR (Partial) Agonist1
Binding Affinity () for α7 nAChR12.3 nM4
Functional Potency () at α7 nAChR2 µM4
Intrinsic Activity (vs. ACh)74%4
Selectivity ( for 5-HT3)140 nM4

IV. Preclinical and Pharmacokinetic Profile

Nelonicline was characterized by pharmacokinetic properties suitable for a CNS-targeted oral therapeutic, including good absorption and extensive metabolism by non-CYP pathways.

Absorption, Distribution, Metabolism, and Excretion (ADME)

Nelonicline was developed as an orally active compound.[4] Clinical pharmacokinetic data from a study in healthy human subjects demonstrated that after a single oral dose, the time to reach maximum plasma concentration () was, on average, 3 hours.[13] As a CNS-targeted agent, it was designed to be brain-permeable, a property that was a key optimization goal during the discovery phase for this class of compounds.[2]

Metabolic Pathways and Elimination

A human mass balance study using a single 100-mg oral dose of radiolabeled nelonicline revealed that the drug undergoes extensive hepatic metabolism before excretion.[14] Unchanged parent drug accounted for only 6.6% of the administered dose recovered in urine, confirming that metabolism is the primary route of clearance.[14]

The two primary metabolic transformations identified were:

  1. Aza-adamantane N-oxidation: This pathway leads to the formation of the major metabolite, M1, which constituted 50.3% of the dose recovered in urine.
  2. Aza-adamantane N-glucuronidation: This pathway produces the second major metabolite, M11, accounting for 19.9% of the urinary dose.[14]

These two metabolites, M1 and M11, were also the major drug-related species circulating in plasma, representing 32.6% and 36.6% of the total drug-related material, respectively. Crucially, these major metabolites were determined to be pharmacologically inactive, with no expected on- or off-target activities, thereby ensuring that the observed clinical effects (or lack thereof) could be attributed to the parent compound.[14]

The enzymes responsible for nelonicline's metabolism are primarily non-cytochrome P450 (CYP) enzymes. The formation of M1 (N-oxidation) is mediated mainly by flavin-containing monooxygenase 3 (FMO3) in the liver, with a minor contribution from FMO1 in the kidney. The formation of M11 (N-glucuronidation) is catalyzed predominantly by UDP-glucuronosyltransferase 1A4 (UGT1A4) and to a lesser extent by UGT2B10.[2] This metabolic profile is highly advantageous from a clinical perspective. Because nelonicline's clearance bypasses the major CYP enzyme system (e.g., CYP3A4, CYP2D6), it has a significantly lower potential for drug-drug interactions. This is a particularly important safety feature for the intended patient populations (elderly individuals with AD and patients with schizophrenia), who are often treated with multiple concurrent medications.

The primary route of elimination for nelonicline and its metabolites is via the kidneys. The human mass balance study showed a total dose recovery of 94.0%, with 81.5% of the administered radioactivity excreted in urine and only 12.4% in feces.[14]

Table 2: Summary of Human Pharmacokinetic (ADME) Parameters for Nelonicline

PK ParameterValue / DescriptionSource(s)
Administration RouteOral4
(Time to Peak Concentration)~3 hours13
Total Dose Recovery94.0%14
Primary Excretion RouteRenal (81.5% of dose in urine)14
Extent of MetabolismExtensive (6.6% of dose as unchanged drug in urine)14
Primary Metabolic Pathway 1Aza-adamantane N-oxidation14
Primary Metabolite 1 (M1)50.3% of dose in urine14
Primary Metabolic Pathway 2Aza-adamantane N-glucuronidation14
Primary Metabolite 2 (M11)19.9% of dose in urine14
Key Metabolizing EnzymesFMO3, UGT1A4, UGT2B102

V. Clinical Development in Alzheimer's Disease

Nelonicline underwent extensive Phase 2 clinical evaluation for the treatment of mild-to-moderate Alzheimer's disease. Despite a strong rationale, the clinical program ultimately failed to demonstrate efficacy, leading to the discontinuation of its development for this indication.[1]

Review of Phase 2 Monotherapy Trials

The initial proof-of-concept for nelonicline as a monotherapy was assessed in trial NCT00948909. This was a 12-week, randomized, double-blind study that compared two doses of nelonicline (5 mg and 25 mg once daily) against both placebo and an active control, donepezil (10 mg), in 274 subjects with mild-to-moderate AD who were not receiving other dementia medications.[15]

The primary endpoint was the change from baseline on the 11-item ADAS-Cog total score. The study did not meet this primary endpoint, as neither dose of nelonicline produced a statistically significant improvement over placebo.[17] The 5 mg dose showed results numerically similar to placebo. However, the 25 mg dose showed a trend toward cognitive improvement that did not reach statistical significance (least squares mean difference from placebo of -1.19; one-sided p=0.095). This trend was comparable to that observed with the active comparator, donepezil (difference from placebo of -1.43; one-sided p=0.057).[17] A subsequent exposure-response analysis of these data revealed a statistically significant relationship between higher plasma concentrations of nelonicline and greater improvement on the ADAS-Cog, with no evidence of a therapeutic plateau. This finding provided a rationale for exploring higher doses in future studies.[17]

Review of Phase 2 Adjunctive Therapy Trials

Based on the encouraging, albeit non-significant, signal from the monotherapy trial, a larger and longer study, NCT01549834, was initiated. This 24-week, randomized, double-blind, placebo-controlled trial evaluated nelonicline as an adjunctive therapy in 434 subjects with mild-to-moderate AD who were already on stable doses of acetylcholinesterase inhibitors.[15] The study tested the previously studied 25 mg dose and a higher 75 mg dose against placebo.

The primary endpoint was again the change from baseline in the ADAS-Cog score, this time at week 24. The study failed to meet its primary endpoint, with neither the 25 mg nor the 75 mg dose of nelonicline demonstrating a significant improvement compared with placebo at the final timepoint. Furthermore, no treatment effect was observed on any of the secondary efficacy measures assessing cognition, function, or global clinical impression.[20] A transient, statistically significant improvement on the ADAS-Cog was noted for the 25 mg dose at the week 4 assessment (p < 0.010), but this effect was not maintained at later timepoints and was therefore not considered clinically meaningful.[20]

Synthesis of Efficacy Findings in AD

The clinical development of nelonicline in AD followed a logical but ultimately unsuccessful path. The initial monotherapy trial provided a weak signal of activity and an exposure-response relationship that suggested higher doses might be effective. This hypothesis was directly tested in the larger, longer adjunctive therapy trial. The definitive failure of this confirmatory study to show any sustained benefit, even at a higher dose, effectively invalidated the initial hypothesis. The final conclusion from the clinical program was that the efficacy profile of nelonicline did not warrant further development for the treatment of mild-to-moderate AD.[20] Several long-term, open-label extension studies associated with the AD program were also subsequently terminated.[2]

Table 3: Summary of Phase 2 Clinical Trials of Nelonicline in Alzheimer's Disease

Trial IdentifierPhaseDesignPopulationDosesPrimary EndpointKey OutcomeSource(s)
NCT009489092Randomized, Double-Blind, Placebo- & Active-Controlled274 subjects with mild-to-moderate AD (monotherapy)5 mg, 25 mgChange in ADAS-Cog at 12 weeksFailed. Trend toward improvement with 25 mg dose (p=0.095).17
NCT015498342Randomized, Double-Blind, Placebo-Controlled434 subjects with mild-to-moderate AD (adjunctive to AChEIs)25 mg, 75 mgChange in ADAS-Cog at 24 weeksFailed. No significant improvement at week 24 for either dose.20

VI. Clinical Development in Schizophrenia

In parallel with the AD program, nelonicline was extensively investigated for its potential to treat Cognitive Impairment Associated with Schizophrenia (CIAS), a core feature of the illness and a major driver of functional disability for which no approved pharmacological treatments exist. The program was designed to rigorously assess efficacy while accounting for the high rate of smoking in this patient population, a key potential confounder.

Efficacy in Cognitive Impairment (CIAS)

Two large Phase 2b studies were conducted to evaluate nelonicline's pro-cognitive effects. The first, NCT01655680, was a 24-week, multicenter trial in 432 clinically stable, non-smoking subjects with schizophrenia.[4] Participants were randomized to receive nelonicline (25 mg, 50 mg, or 75 mg once daily) or placebo as an add-on to their existing antipsychotic medication. The primary endpoint was the change from baseline to week 12 on the MATRICS Consensus Cognitive Battery (MCCB) neurocognitive composite score. The study failed to meet its primary endpoint, with no statistically significant differences observed between any nelonicline dose and placebo on the MCCB score.[22]

The second major study, NCT01678755, employed a similar design but focused on a population of 157 smokers with stable schizophrenia.[12] This 12-week trial compared nelonicline (25 mg and 75 mg) with placebo. It also failed to meet its primary endpoint, demonstrating no pro-cognitive effect on the MCCB neurocognitive composite score in this population.[12]

The consistent failure to improve cognition across both non-smoking and smoking populations provided a robust and definitive negative result. This outcome strongly suggests that the therapeutic strategy of direct α7 nAChR agonism is not sufficient to ameliorate the complex cognitive deficits inherent to schizophrenia. The failure in both groups, despite the careful trial designs, indicates that the initial hypothesis—that patients smoke to self-medicate cognitive deficits via nicotinic stimulation and that a selective agonist could replicate this effect therapeutically—was likely overly simplistic.

Observed Effects on Negative Symptoms

Despite the failure to meet the primary cognitive endpoints, an unexpected and intriguing secondary finding emerged from both schizophrenia trials. In the non-smoker study (NCT01655680), a trend toward improvement in negative symptoms was observed at week 24 for the 50 mg dose, as measured by the 16-item Negative Symptom Assessment (NSA-16) total score (change from baseline of -4.27 vs. -3.00 for placebo; p=0.059).[4] A similar finding was reported in the smoker study (NCT01678755), where the 75 mg dose showed a trend for improvement on the NSA-16 total score at week 12 (p=0.053).[12]

The replication of this weak but consistent signal across two large, independent studies suggests it may represent a real, albeit modest, pharmacological effect of nelonicline on the negative symptom domain of schizophrenia (e.g., avolition, anhedonia, social withdrawal). While this effect was not strong enough to warrant continued development of the drug, it provides valuable scientific information, hinting that α7 nAChR modulation might influence neurotransmitter systems relevant to negative symptoms. This finding could potentially inform future drug discovery efforts targeting this notoriously difficult-to-treat aspect of schizophrenia.

Table 4: Summary of Phase 2 Clinical Trials of Nelonicline in Schizophrenia

Trial IdentifierPhaseDesignPopulationDosesPrimary EndpointKey Cognitive OutcomeKey Negative Symptom OutcomeSource(s)
NCT016556802bRandomized, Double-Blind, Placebo-Controlled432 non-smoking subjects with stable schizophrenia25, 50, 75 mgChange in MCCB at 12 weeksFailed. No significant difference vs. placebo.Trend for improvement on NSA-16 with 50 mg dose (p=0.059).22
NCT016787552bRandomized, Double-Blind, Placebo-Controlled157 smoking subjects with stable schizophrenia25, 75 mgChange in MCCB at 12 weeksFailed. No significant difference vs. placebo.Trend for improvement on NSA-16 with 75 mg dose (p=0.053).12

VII. Consolidated Safety and Tolerability Assessment

A consistent finding across the entire Phase 2 clinical development program for nelonicline was its favorable safety and tolerability profile. In studies involving patients with both Alzheimer's disease and schizophrenia, the drug was reported to be generally well tolerated.[9]

The overall incidence and types of adverse events (AEs) associated with nelonicline treatment were consistently described as being similar to placebo.[12] In the AD monotherapy trial, no clinically meaningful differences in safety measures were observed between the nelonicline groups, placebo, and the active comparator, donepezil.[17] The severity of the vast majority of reported AEs was mild to moderate, and there was no evidence that nelonicline increased the frequency of severe or serious adverse events compared to placebo.[18]

The most frequently reported treatment-emergent adverse events across the clinical program included [12]:

  • Gastrointestinal: Diarrhea, nausea, and constipation. In the study of smokers with schizophrenia (NCT01678755), constipation was the only AE with a notably higher incidence in the combined nelonicline groups (5.8%) compared to the placebo group (0%).[12]
  • Neurological: Dizziness and headache.
  • General: Fatigue, nasopharyngitis, agitation, and falls (reported in the AD population).

The clean safety profile of nelonicline is a critical aspect of its development story. It demonstrates that the program was not halted due to unacceptable toxicity, which can sometimes prevent drugs from being tested at potentially efficacious doses. In the case of nelonicline, the drug was tested at a range of doses, including those predicted to be effective based on exposure-response modeling, and was found to be safe. This removes any ambiguity regarding the reason for its discontinuation, isolating the cause to a clear and definitive lack of clinical benefit.

VIII. Conclusion: Developmental Trajectory and Future Perspectives

The development of nelonicline (ABT-126) represents a well-conceived and rigorously executed effort to validate the α7 nicotinic acetylcholine receptor as a therapeutic target for treating cognitive dysfunction in Alzheimer's disease and schizophrenia. The program successfully advanced a molecule with many desirable drug-like properties, including target selectivity, oral bioavailability, CNS penetration, and a favorable safety profile with a low propensity for drug-drug interactions.

Despite this strong foundation, the comprehensive Phase 2 clinical program yielded unequivocally negative results regarding the primary efficacy endpoints. In multiple large, placebo-controlled trials, nelonicline failed to demonstrate a statistically significant or clinically meaningful improvement in cognition in either patient population.[7] The ultimate decision to discontinue the development of nelonicline was therefore a direct consequence of this lack of demonstrable efficacy.[1] The conclusion from the clinical data was clear: the drug's efficacy profile did not warrant the significant investment required for Phase 3 development.[20]

The failure of nelonicline, alongside other α7 nAChR agonists like encenicline, has had important implications for the field. It highlights a significant translational gap, where promising and robust effects in preclinical animal models did not translate to efficacy in human trials.[12] This outcome has called into question the viability of the direct orthosteric agonist mechanism for a receptor known for its rapid and profound desensitization. The experience with nelonicline suggests that more nuanced approaches to modulating the α7 nAChR, such as positive allosteric modulation, may be necessary to achieve a therapeutic effect without triggering the receptor's intrinsic negative feedback mechanisms. While the program did not achieve its primary goal, the unexpected and replicated signal on negative symptoms in schizophrenia provides a valuable scientific observation that may inform future research into the diverse roles of the cholinergic system in psychiatric illness.

Works cited

  1. Nelonicline - AbbVie - AdisInsight - Springer, accessed October 12, 2025, https://adisinsight.springer.com/drugs/800029971
  2. Nelonicline - Drug Targets, Indications, Patents - Patsnap Synapse, accessed October 12, 2025, https://synapse-patsnap-com.libproxy1.nus.edu.sg/drug/26908cd4ddf04b7b9bd601e51d6ff046
  3. Abt-126 | C17H19N3OS | CID 24987875 - PubChem, accessed October 12, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Abt-126
  4. Nelonicline (ABT-126) | α7 nAChR Agonist | MedChemExpress, accessed October 12, 2025, https://www.medchemexpress.com/Nelonicline.html
  5. Nelonicline |CAS:1026134-63-3 Probechem Biochemicals, accessed October 12, 2025, https://www.probechem.com/products_Nelonicline.html
  6. Nelonicline: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed October 12, 2025, https://go.drugbank.com/drugs/DB19040
  7. Nicotinic Acetylcholine Receptor Agonists for the Treatment of Alzheimer's Dementia: An Update - PMC - PubMed Central, accessed October 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6379052/
  8. Nelonicline - Sobekbio Biosciences, accessed October 12, 2025, https://sobekbio.com/suppliers/molnova/product/624206-nelonicline-1
  9. Alzheimer's Disease: Dawn of a New Era? - Publishing at the Library, accessed October 12, 2025, https://journals.library.ualberta.ca/jpps/index.php/JPPS/article/download/29185/21354/77856
  10. A Systematic Review on Drugs Acting as Nicotinic Acetylcholine Receptor Agonists in the Treatment of Dementia - PubMed Central, accessed October 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10854606/
  11. The History of Alzheimer's Disease - BrightFocus Foundation, accessed October 12, 2025, https://www.brightfocus.org/resource/the-history-of-alzheimers-disease/
  12. Efficacy and Safety of the α7-Nicotinic Acetylcholine Receptor Agonist ABT-126 in the Treatment of Cognitive Impairment Associated With Schizophrenia: Results From a Phase 2b Randomized Controlled Study in Smokers - ResearchGate, accessed October 12, 2025, https://www.researchgate.net/publication/319651786_Efficacy_and_Safety_of_the_a7-Nicotinic_Acetylcholine_Receptor_Agonist_ABT-126_in_the_Treatment_of_Cognitive_Impairment_Associated_With_Schizophrenia_Results_From_a_Phase_2b_Randomized_Controlled_Stud
  13. NELONICLINE N-OXIDE - precisionFDA, accessed October 12, 2025, https://precision.fda.gov/ginas/app/ui/substances/0ba9c0b2-11e2-4b48-9767-926bdf8a525f
  14. Metabolism and Disposition of a Novel Selective α 7 Neuronal ..., accessed October 12, 2025, https://pubmed.ncbi.nlm.nih.gov/29348125/
  15. Nelonicline Completed Phase 2 Trials for Alzheimer's Disease (AD ..., accessed October 12, 2025, https://go.drugbank.com/drugs/DB19040/clinical_trials?conditions=DBCOND0049114&phase=2&purpose=treatment&status=completed
  16. NCT00948909 | Efficacy and Safety Study for Subjects With Mild-to-Moderate Alzheimer's Disease | ClinicalTrials.gov, accessed October 12, 2025, https://clinicaltrials.gov/study/NCT00948909?term=NCT00867399%20NCT00948909%20NCT01482845%20NCT01527916%20NCT01549834%20NCT01676935%20NCT01690195&rank=6
  17. A phase 2 randomized, controlled trial of the α7 agonist ABT-126 in mild-to-moderate Alzheimer's dementia - PubMed, accessed October 12, 2025, https://pubmed.ncbi.nlm.nih.gov/29854928/
  18. A phase 2 randomized, controlled trial of the α7 agonist ABT-126 in mild-to-moderate Alzheimer's dementia - ResearchGate, accessed October 12, 2025, https://www.researchgate.net/publication/280925518_A_phase_2_randomized_controlled_trial_of_the_a7_agonist_ABT-126_in_mild-to-moderate_Alzheimer's_dementia
  19. NCT01549834 - Patients at Heart, accessed October 12, 2025, https://patientsatheart.com/trials/nct01549834
  20. Efficacy and Safety of ABT-126 in Subjects with Mild-to-Moderate Alzheimer's Disease on Stable Doses of Acetylcholinesterase Inhibitors: A Randomized, Double-Blind, Placebo-Controlled Study - ResearchGate, accessed October 12, 2025, https://www.researchgate.net/publication/297900768_Efficacy_and_Safety_of_ABT-126_in_Subjects_with_Mild-to-Moderate_Alzheimer's_Disease_on_Stable_Doses_of_Acetylcholinesterase_Inhibitors_A_Randomized_Double-Blind_Placebo-Controlled_Study
  21. NCT01655680 | A Study to Evaluate ABT-126 for the Treatment of Cognitive Deficits in Schizophrenia | ClinicalTrials.gov, accessed October 12, 2025, https://clinicaltrials.gov/study/NCT01655680
  22. The α7 Nicotinic Agonist ABT-126 in the Treatment of Cognitive Impairment Associated with Schizophrenia in Nonsmokers: Results from a Randomized Controlled Phase 2b Study - PubMed, accessed October 12, 2025, https://pubmed.ncbi.nlm.nih.gov/27319970/
  23. NCT01678755 | A Phase 2 Study to Evaluate ABT-126 for the Treatment of Cognitive Deficits in Schizophrenia | ClinicalTrials.gov, accessed October 12, 2025, https://clinicaltrials.gov/study/NCT01678755
  24. Efficacy and Safety of the α7-Nicotinic Acetylcholine Receptor Agonist ABT-126 in the Treatment of Cognitive Impairment Associated With Schizophrenia: Results From a Phase 2b Randomized Controlled Study in Smokers - PubMed, accessed October 12, 2025, https://pubmed.ncbi.nlm.nih.gov/28922590/
  25. nelonicline (ABT-126) / AbbVie - LARVOL DELTA, accessed October 12, 2025, https://delta.larvol.com/Products/?ProductId=fe7c63ea-2d77-48fa-b9ce-03c62f987970
  26. Alzheimer's Disease - Publishing at the Library, accessed October 12, 2025, https://journals.library.ualberta.ca/jpps/index.php/JPPS/article/viewFile/29185/21354
  27. 32.2 TWO GLOBAL PHASE III TRIALS OF ENCENICLINE FOR, accessed October 12, 2025, https://www.researchgate.net/publication/332308614_322_TWO_GLOBAL_PHASE_III_TRIALS_OF_ENCENICLINE_FOR_COGNITIVE_IMPAIRMENT_IN_CHRONIC_SCHIZOPHRENIA_PATIENTS_RED_FLAGS_AND_LESSONS_LEARNED

Published at: October 12, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.