MedPath

Amlenetug Advanced Drug Monograph

Published:May 14, 2025

Generic Name

Amlenetug

Amlenetug (Lu AF82422): An Investigational Monoclonal Antibody for Multiple System Atrophy

1. Executive Summary

Amlenetug, also identified by its investigational name Lu AF82422, is a human monoclonal antibody currently under development for the treatment of Multiple System Atrophy (MSA), a rare, progressive, and ultimately fatal neurodegenerative disorder for which no disease-modifying therapies are currently approved.[1] Developed by H. Lundbeck A/S in collaboration with Genmab A/S, Amlenetug targets extracellular α-synuclein, a protein centrally implicated in the pathology of MSA and other synucleinopathies.[1] The therapeutic rationale is to inhibit the uptake and aggregation of pathological α-synuclein, thereby slowing disease progression.

The Phase 2 AMULET trial (NCT05104476), while not meeting its primary efficacy endpoint in the overall MSA patient population, demonstrated a 19% slowing of clinical progression on the Unified MSA Rating Scale (UMSARS) in the active treatment arm compared to placebo, and a more pronounced, albeit non-significant, 37% slowing of clinical progression in a pre-specified subgroup of less-impaired patients.[12] Encouraged by these signals and a generally well-tolerated safety profile, Lundbeck has advanced Amlenetug into a global Phase 3 clinical trial, MASCOT (NCT06706622).[1] The development program has received significant regulatory support, including Orphan Drug Designation in the US, EU, and Japan, and Fast Track Designation from the US FDA, underscoring the high unmet medical need in MSA.[6]

The decision to proceed to a large-scale Phase 3 trial, despite the Phase 2 study not achieving statistical significance on its primary endpoint in the overall cohort, is a notable aspect of Amlenetug's development. This progression reflects the critical unmet medical need in MSA, a disease with no approved treatments that can alter its course.[1] In such scenarios, particularly in rare diseases, regulatory agencies and pharmaceutical developers may pursue therapeutic candidates that show promising signals of efficacy in specific patient subgroups, provided the safety profile is acceptable. The 37% slowing of disease progression observed in the less-impaired subgroup within the AMULET trial [12], coupled with acceptable tolerability, likely provided a compelling rationale for further investigation in a larger, potentially more focused, Phase 3 study. This approach acknowledges the inherent difficulties in demonstrating efficacy in heterogeneous neurodegenerative populations and the ethical imperative to explore promising avenues for devastating conditions.

Furthermore, Amlenetug's mechanism of action, which targets the accumulation and spread of α-synuclein, aligns with a central hypothesis regarding the pathogenesis of MSA and other synucleinopathies like Parkinson's disease.[4] The success or failure of Amlenetug in Phase 3 will therefore not only determine its future as a potential therapy for MSA but also contribute significantly to the broader understanding and validation of the α-synuclein hypothesis as a therapeutic target in these debilitating neurological conditions.

2. Introduction to Amlenetug and Multiple System Atrophy (MSA)

Overview of Multiple System Atrophy (MSA)

Multiple System Atrophy (MSA) is a rare, adult-onset, sporadic neurodegenerative disorder characterized by a relentlessly progressive course and a fatal outcome.[1] Clinically, MSA presents with a variable combination of parkinsonism (MSA-P subtype), cerebellar ataxia (MSA-C subtype), autonomic failure (such as cardiovascular and urogenital dysfunction), and pyramidal signs.[4] The typical age of onset is between 55 and 60 years.[4] Epidemiological studies estimate the prevalence of MSA to be between 1.9 and 4.9 cases per 100,000 people worldwide, with an incidence in the US of approximately 0.6 per 100,000 person-years, increasing to 3 per 100,000 person-years in individuals aged 50 and older.[4] Despite its devastating impact, there are currently no approved therapies that can slow or halt the underlying neurodegenerative process, representing a significant unmet medical need.[1] Current management is purely symptomatic and supportive.

Amlenetug: Introduction, Chemical Nature, and Developers

Amlenetug, also known by its investigational designation Lu AF82422 (CAS Number: 2850367-41-6), is an investigational therapeutic agent.[10] It is a fully human monoclonal antibody (mAb) of the IgG1 subclass.[1] Amlenetug is being developed by the global pharmaceutical company H. Lundbeck A/S [1] through a joint research and licensing agreement with Genmab A/S, a biotechnology company specializing in antibody therapeutics.[1] As an investigational compound, Amlenetug has not yet received marketing approval from any regulatory authority worldwide.[1]

Table 1: Amlenetug - Key Drug Characteristics

CharacteristicDetail
Generic Name(s)Amlenetug, Lu AF82422
CAS Number2850367-41-6 21
Drug ClassHuman Monoclonal Antibody (IgG1) 1
Developer(s)H. Lundbeck A/S 1
Originator/CollaborationInvented by Lundbeck in collaboration with Genmab A/S 1
Therapeutic TargetExtracellular α-synuclein (all major forms, with high affinity for oligomers) 1
Primary IndicationMultiple System Atrophy (MSA) 1

This table provides a foundational summary of Amlenetug, consolidating key identifiers and developmental context crucial for understanding its profile.

3. Mechanism of Action and Preclinical Rationale

Targeting Alpha-Synuclein in Neurodegeneration

Alpha-synuclein (α-syn) is a 15 kDa protein predominantly expressed in neurons and also found in peripheral nerves and blood cells.[7] While its precise physiological functions are still being elucidated, it is thought to play roles in synaptic vesicle trafficking and neurotransmitter release.[28] In several neurodegenerative disorders, collectively termed synucleinopathies, including MSA, Parkinson's disease (PD), and Dementia with Lewy Bodies, α-syn misfolds and aggregates into insoluble pathological forms such as oligomers, protofibrils, and fibrils, which are the main components of characteristic intracellular inclusions like Lewy bodies and glial cytoplasmic inclusions.[4] The accumulation and cell-to-cell propagation of these toxic α-syn species are believed to be central to the progressive neuronal dysfunction and degeneration observed in these conditions.[4] Consequently, therapeutic strategies aimed at reducing the burden of pathological α-syn, inhibiting its aggregation, or preventing its spread are considered promising approaches for disease modification.[7] Immunotherapy, both active (vaccination) and passive (monoclonal antibody administration), targeting these pathological α-syn species is a key area of investigation.[7]

Amlenetug's Specific Mechanism

Amlenetug is a human IgG1 monoclonal antibody specifically engineered to target extracellular forms of α-synuclein.[1] It is designed to recognize and bind to all major species of extracellular α-syn, reportedly with high affinity for oligomeric forms, which are considered particularly neurotoxic.[7] The proposed therapeutic mechanism of Amlenetug is multifaceted:

  1. Prevention of Uptake and Seeding: By binding to extracellular pathological α-syn aggregates, Amlenetug aims to prevent their uptake by neighboring neurons and glial cells. This is crucial as the intercellular "seeding" and spread of misfolded α-syn is thought to drive the progression of pathology throughout the brain.[1]
  2. Inhibition of Aggregation: Amlenetug is intended to inhibit the further aggregation of α-synuclein.[1]
  3. Immune-Mediated Clearance: Amlenetug possesses an active Fc region. This component of the antibody can engage Fc receptors on immune cells, particularly microglia (the resident immune cells of the brain). This engagement is hypothesized to facilitate the clearance of α-synuclein/Amlenetug complexes from the extracellular space via microglial phagocytosis.[4]

This comprehensive approach—neutralizing extracellular α-syn, preventing its pathogenic spread, and promoting its removal—forms the basis of Amlenetug's potential as a disease-modifying therapy for MSA. The ability to not only bind but also to potentially inhibit propagation and enhance clearance through the active Fc region suggests a strategy designed to intervene at multiple points in the α-synuclein pathogenic cascade.

Summary of Key Preclinical Findings

The development of Amlenetug is supported by several preclinical findings. In vitro studies using tissue from MSA patients demonstrated that Lu AF82422 binds to aggregated α-synuclein isolated from human MSA brain tissue with an EC50 of 0.09 nM and inhibits the seeding activity of this pathological α-syn.[4] This direct evidence of interaction with human disease-relevant pathology is a significant preclinical validation.

Pharmacokinetic studies in cynomolgus monkeys with single doses of Lu AF82422 (1-30 mg/kg) confirmed PK properties typical for a monoclonal antibody with a soluble target, and target engagement was demonstrated by a decrease in free α-synuclein in plasma.[7] Furthermore, four-week repeat-dose intravenous toxicity studies in rats and cynomolgus monkeys, with doses up to 600 mg/kg administered every 10 days, showed no treatment-related adverse findings. The no-observed-adverse-effect-level (NOAEL) was established at the highest dose tested (600 mg/kg), providing an adequate safety margin for progression to human clinical trials. Target engagement was also shown in plasma and cerebrospinal fluid (CSF) in these animal studies.[7]

Flow cytometry analysis indicated that Lu AF82422 did not bind to the extracellular surface of human platelets, erythrocytes, granulocytes, or lymphocytes, but did show low-level binding to a small fraction of monocytes, without any observed functional consequences on their activation or phagocytic capacity.[7] A tissue cross-reactivity assessment showed that Lu AF82422 binding was generally restricted to nervous tissues.[7] Collectively, these nonclinical data indicated no safety signals of concern and supported the initiation of clinical trials.[7] The ability of the antibody to engage its target in the CSF of animal models, and later confirmed in human PD patients [7], is particularly important, as it suggests Amlenetug can cross the blood-brain barrier to reach pathological α-synuclein within the central nervous system, a critical factor for potential efficacy in neurodegenerative diseases like MSA.

4. Clinical Development Program

The clinical development of Amlenetug has progressed through Phase 1 and Phase 2 studies, with a Phase 3 trial currently underway.

Table 2: Overview of Amlenetug Clinical Trials

Trial ID (NCT)PhaseTitle/Brief DescriptionStatus (as of latest info)Patient PopulationN (Approx.)Basic DesignKey Objectives
NCT03611569 171A Trial of Lu AF82422 in Healthy Non-Japanese and Japanese Subjects and in Patients With Parkinson's Disease 17Completed (July 2021) 20Healthy volunteers (Japanese & Non-Japanese); Parkinson's Disease patients 1759 healthy, 15 PD 17Randomized, placebo-controlled, ascending single dose 29Safety, tolerability, PK, target engagement 17
NCT05104476 12AMULET: A Trial of Lu AF82422 in Participants With Multiple System Atrophy 1Active, not recruiting (Dec 2022); OLE ongoing 1Multiple System Atrophy (MSA) patients 161 1Randomized (2:1), double-blind, placebo-controlled, OLE 1Efficacy (UMSARS progression), safety, tolerability 1
NCT06706622 63MASCOT: A Trial of Lu AF82422 in Participants With Multiple System Atrophy 6Active, Recruiting (as of Feb 2025) 14Multiple System Atrophy (MSA-P or MSA-C) patients 6>300 13Randomized, double-blind, placebo-controlled (high/low dose Amlenetug vs placebo), OLE 1Efficacy, safety, tolerability 1
NCT06258720 231A Trial to Assess Safety, Tolerability, Pharmacokinetics, and Immunogenicity of Lu AF82422 in Healthy Caucasian and Chinese Adult Participants 23Active, Recruiting (estimated completion Sep 2024) 23Healthy Caucasian and Chinese adults 23Not specifiedSingle infusion 23Safety, tolerability, PK, immunogenicity 23

This table summarizes the progression of Amlenetug's clinical investigation, highlighting the different populations and study designs employed at each stage.

Phase 1 Studies

NCT03611569 (Healthy Volunteers and PD Patients)

The first-in-human study (NCT03611569) was a randomized, placebo-controlled, ascending single-dose trial designed to evaluate the safety, tolerability, pharmacokinetics (PK), and target engagement of Lu AF82422.[17] The study enrolled two main cohorts: Cohort A consisted of healthy participants aged 18-55 years (further divided into non-Japanese and Japanese subjects), and Cohort B comprised patients with Parkinson's disease (PD) aged 40-80 years with Hoehn and Yahr stage ≤3.[17] Participants received single intravenous (IV) infusions of Lu AF82422 at doses ranging from 75 mg to 9000 mg, or placebo, and were monitored for a 12-week observational period.[16]

Key results from this Phase 1 trial, published by Buur et al. in Movement Disorders (2024), indicated that single IV infusions of Lu AF82422 were generally safe and well-tolerated across all dose levels in both healthy volunteers and PD patients.[7] No serious adverse events (AEs) were reported.[7] The most commonly observed AEs were related to study procedures such as lumbar puncture, headache, and common infections, and were mostly mild and transient.[7]

Pharmacokinetic analysis revealed that Lu AF82422 concentrations in plasma and cerebrospinal fluid (CSF) increased in a dose-proportional manner, with no observable differences between cohorts.[7] The mean plasma half-life of Lu AF82422 was approximately 700 hours.[7] Pharmacodynamic assessments demonstrated target engagement. Plasma concentrations of Lu AF82422 had an immediate, concentration-dependent lowering effect on the plasma concentration of free α-synuclein and on the ratio of free-to-total α-synuclein in all cohorts.[7] Importantly, in the high-dose PD cohort (9000 mg), Lu AF82422 also lowered the free-to-total α-synuclein ratio in the CSF, indicating central target engagement.[7] These findings supported the further clinical development of Lu AF82422.[7]

NCT06258720 (Healthy Caucasian and Chinese Adults)

A separate Phase 1 trial (NCT06258720) was registered to assess the safety, tolerability, pharmacokinetics, and immunogenicity of a single infusion of Lu AF82422 in healthy Caucasian and Chinese adult participants.[23] This study was reported as active and recruiting, with an estimated completion date of September 2024.[23] This trial suggests a strategic approach to understanding potential ethnic differences, which can be important for global drug development and regulatory submissions in diverse regions.

Phase 2 AMULET Trial (NCT05104476) in MSA

Trial Design and Patient Population

The AMULET study (NCT05104476) was a Phase 2, randomized, double-blind, placebo-controlled, multicenter clinical trial designed to evaluate the efficacy and safety of Amlenetug in patients with MSA.[1] The trial was conducted at 21 sites in the USA and Japan.[4] A total of 61 participants with a diagnosis of possible or probable MSA (either MSA-P or MSA-C subtypes) were enrolled.[1] Key inclusion criteria included age between 40 and 75 years, onset of motor symptoms within the last 5 years, an anticipated survival of at least 3 years, a Unified MSA Rating Scale (UMSARS) Part I score of ≤16 (omitting item 11, which pertains to sexual function), and a Montreal Cognitive Assessment (MoCA) score of ≥22.[4] Participants were randomized in a 2:1 ratio to receive either Amlenetug or placebo.[1]

Dosage and Administration

Participants in the active arm received Amlenetug at a dose of 4.2 g, administered as an intravenous (IV) infusion every four weeks (Q4W).[1] The double-blind treatment period lasted between 48 and 72 weeks.[1]

Primary and Secondary Endpoints

The primary objective of the AMULET trial was to evaluate the efficacy of Amlenetug on clinical progression in patients with MSA.[1] This was assessed using a Bayesian progression model of the longitudinal changes from baseline in the Total UMSARS score (sum of Part I and Part II) up to Week 72, aiming to show a slowing in clinical progression in the active treatment arm compared to placebo on a 5% significance level (1-sided).[1] Secondary objectives included the evaluation of Amlenetug's effects on patient functioning, disease severity, and other aspects of MSA, such as modified UMSARS, UMSARS Part I, and UMSARS Part II scores.[1] Safety and tolerability were also key endpoints.[1]

Efficacy Results

Table 3: Summary of Efficacy Results from AMULET (Phase 2) Trial

EndpointPopulationAmlenetug Group Result (% slowing vs. placebo)Statistical Significance
Total UMSARS (Part I+II) Score (Primary Endpoint)Overall Population (N=61)19% slowing 12Primary endpoint not met (non-significant) 13
Total UMSARS (Part I+II) ScoreLess Impaired Subgroup (UMSARS Part I ≤16 at baseline; n=30 Amlenetug, n=12 Placebo)37% slowing 12Not specified as primary for subgroup; trend observed
Modified UMSARS (Key Secondary)Overall Population27% slowing 15Trend observed
UMSARS Part IOverall Population22% slowing 15Trend observed
UMSARS Part IIOverall Population17% slowing 15Trend observed

The AMULET trial did not meet its primary endpoint in the overall study population, as the observed 19% slowing of clinical progression on the Total UMSARS score in the Amlenetug arm compared to placebo was not statistically significant.[12] However, a pre-specified subgroup analysis of less-impaired participants (defined by a baseline UMSARS Part I score ≤16) showed a more pronounced effect. In this subgroup, consisting of 30 patients treated with Amlenetug and 12 with placebo, Amlenetug treatment was associated with a 37% slowing of clinical progression compared to placebo.[12] Consistent trends were also observed in analyses of the modified UMSARS (a key secondary endpoint, showing 27% slowing), UMSARS Part I (22% slowing), and UMSARS Part II (17% slowing) in the overall population.[15] These findings, particularly from the less-impaired subgroup, suggested a potential for Amlenetug to modify disease progression, especially if initiated earlier in the disease course.

Safety and Tolerability Profile

Table 4: Summary of Safety Profile of Amlenetug (from AMULET and Phase 1 data)

TrialMost Common Adverse Events (AEs)Serious Adverse Events (SAEs)Discontinuation Rates due to AEs
Phase 1 (NCT03611569) 7Lumbar puncture-related effects, headache, common infectionsNone reported as serious in abstractsNot specified, but no withdrawals due to AEs in some reports
Phase 2 AMULET (NCT05104476) 15Generally well tolerated (specific AEs not detailed in provided abstracts)Not detailed in provided abstractsNot detailed in provided abstracts, but 45/61 continued to OLE

Amlenetug was reported to be generally well-tolerated in the AMULET trial.[15] The decision of 45 out of the 61 participants to continue into the open-label extension phase further supports its acceptable safety and tolerability profile, or at least a perceived favorable benefit-risk balance by patients and investigators.[13]

Open-Label Extension (OLE) Phase

Following the 48-72 week double-blind treatment period, participants in the AMULET trial had the option to enter an ongoing 96-week open-label extension (OLE) phase, during which all participants are offered treatment with Amlenetug.[1] This OLE phase is crucial for gathering long-term safety and efficacy data.

Phase 3 MASCOT Trial (NCT06706622) in MSA

Rationale and Objectives

The initiation of the Phase 3 MASCOT (Multiple System Atrophy α-Synuclein COrrection Trial) study was based on the totality of data from the AMULET trial, particularly the encouraging signals of efficacy observed in the less-impaired patient subgroup, and the drug's favorable safety profile.[1] The primary objective of the MASCOT trial is to definitively evaluate the efficacy and safety of Amlenetug in slowing disease progression in patients with MSA and to confirm the potential effects on clinical progression suggested by the Phase 2 data.[15]

Trial Design, Patient Population, and Endpoints

MASCOT (NCT06706622) is a global, Phase 3, interventional, randomized, double-blind, parallel-group, placebo-controlled trial with an optional open-label extension.[1] The trial plans to enroll over 300 participants with MSA, including both parkinsonian (MSA-P) and cerebellar (MSA-C) subtypes, across multiple sites in North America, Europe, Asia, and Australia.[3]

The trial comprises two parts:

  1. A 72-week double-blind treatment period where participants are randomized to receive either a high dose of Amlenetug, a low dose of Amlenetug, or placebo.[1]
  2. An optional open-label extension period where all participants enrolled in the trial will be offered treatment with Amlenetug.[1]

Amlenetug will be administered via intravenous infusion every four weeks.[1] Key eligibility criteria for the MASCOT trial include a diagnosis of clinically established or probable MSA according to the 2022 Movement Disorders Society (MDS) criteria, onset of motor symptoms within the past five years, an anticipated survival of over three years, and a UMSARS Part I score of ≤16 (excluding the sexual function item).[16] Exclusion criteria include prior exposure to Amlenetug, recent use of other investigational drugs, a family history of MSA in two or more first-degree relatives, unexplained anosmia in MSA-P, or other conditions that could impair CNS function or mimic MSA symptoms.[16] The primary endpoint will likely focus on change in UMSARS scores to assess disease progression.

Current Status

The MASCOT trial was reported to have been initiated in November 2024 [16], with other sources indicating a start in December 2024 [20] or more generally in early 2025.[6] As of February 2025, the trial is active and recruiting participants.[14]

The clinical development of Amlenetug illustrates a data-driven, adaptive approach. The progression from a broader Phase 1 population including PD patients to a specific MSA focus in Phase 2, and the subsequent refinement of the target population for Phase 3 based on subgroup analyses from AMULET (particularly focusing on less impaired patients [12]), highlights this strategy. The use of Bayesian modeling for UMSARS assessment in AMULET [12] also points to efforts to employ sensitive methodologies for detecting treatment effects in a challenging, slowly progressive neurodegenerative disease. The consistent inclusion of OLE phases in both AMULET and MASCOT is a standard and ethically important feature in rare disease trials, allowing continued patient access to potentially beneficial therapy and facilitating the collection of valuable long-term safety and efficacy data.[1] The high rate of continuation from the double-blind phase to the OLE in AMULET (45 out of 61 participants) [13] may suggest a perceived benefit or acceptable tolerability by both patients and investigators.

5. Pharmacokinetics and Pharmacodynamics (PK/PD)

Pharmacokinetic and pharmacodynamic data for Amlenetug primarily derive from the Phase 1 study (NCT03611569) conducted in healthy volunteers and patients with Parkinson's disease.[7] Following single intravenous infusions, Amlenetug concentrations in plasma and cerebrospinal fluid (CSF) were observed to increase in a dose-proportional manner across the tested dose range (75 mg to 9000 mg).[7] The mean plasma half-life of Amlenetug was determined to be approximately 700 hours [7], supporting the every-four-week (Q4W) dosing interval implemented in the Phase 2 and Phase 3 trials for MSA.[1]

Pharmacodynamic assessments demonstrated clear target engagement. Amlenetug administration led to an immediate and concentration-dependent reduction in the plasma levels of free α-synuclein.[7] Furthermore, a decrease in the ratio of free-to-total α-synuclein was observed in the plasma of all participant cohorts.[7] Critically for a CNS-targeting therapeutic, in the high-dose PD patient cohort (9000 mg), Amlenetug also lowered the free-to-total α-synuclein ratio in the CSF.[7]

The demonstration of central target engagement, evidenced by the modulation of α-synuclein ratios in the CSF, is a pivotal finding from the Phase 1 study. For antibody-based therapies aimed at neurodegenerative diseases, achieving sufficient penetration across the blood-brain barrier and interaction with the target protein within the CNS is a major challenge. The observed changes in CSF α-synuclein species suggest that Amlenetug can indeed access the CNS and exert its biological effects on its target, which is fundamental for its potential efficacy in MSA. This, combined with its long plasma half-life that allows for convenient monthly dosing, underpins the rationale for its continued clinical investigation.

6. Regulatory Landscape and Designations

Amlenetug has received several significant regulatory designations from major health authorities worldwide, reflecting the high unmet medical need in MSA and the potential of this investigational therapy.

Table 5: Regulatory Designations for Amlenetug

Designation TypeRegulatory BodyDate Granted (Approximate)Indication
Orphan Drug Designation (ODD)US Food and Drug Administration (FDA)April 2024 9Multiple System Atrophy (MSA)
Orphan Drug Designation (ODD)European Medicines Agency (EMA)May 2021 6Multiple System Atrophy (MSA)
Orphan Drug Designation (ODD)Ministry of Health, Labor and Welfare (MHLW), JapanMarch 2025 68Multiple System Atrophy (MSA)
Fast Track Designation (FTD)US Food and Drug Administration (FDA)February 2025 9Multiple System Atrophy (MSA)
SAKIGAKE DesignationMinistry of Health, Labor and Welfare (MHLW), JapanMarch 2023 6Multiple System Atrophy (MSA)

Orphan Drug Designation (ODD)

Amlenetug has been granted ODD for the treatment of MSA by the FDA in April 2024 [9], the EMA in May 2021 [6], and Japan's MHLW by March 2025.[6] ODD is granted to drugs intended for the treatment, prevention, or diagnosis of rare diseases or conditions (affecting fewer than 200,000 people in the US, or not more than 5 in 10,000 in the EU).[9] This designation provides various incentives to the developer, such as market exclusivity for a defined period post-approval, fee reductions for regulatory activities, and enhanced regulatory support and scientific advice.[6]

Fast Track Designation (FTD)

In February 2025, the FDA granted Fast Track Designation to Amlenetug for its potential as a treatment for MSA.[9] This designation is intended to facilitate the development and expedite the review of drugs that treat serious conditions and fill an unmet medical need.[9] Benefits of FTD include more frequent meetings with the FDA, eligibility for accelerated approval and priority review if relevant criteria are met, and the possibility of a rolling review, where a company can submit completed sections of its Biologic License Application (BLA) or New Drug Application (NDA) for review by FDA, rather than waiting until every section is complete.[9] The FTD for Amlenetug was based on the outcome of the Phase 2 AMULET trial.[9]

SAKIGAKE Designation (Japan)

Amlenetug received SAKIGAKE designation from Japan's MHLW in March 2023.[6] The SAKIGAKE Designation System promotes research and development in Japan of innovative new medicines that are intended for severe and life-threatening diseases, show prominent effectiveness, and are developed initially in Japan or for which an application is planned to be submitted in Japan ahead of other countries. This designation offers benefits such as prioritized consultation, a dedicated review system, and a shortened review period.

Implications of Designations

The collective granting of these special regulatory designations by major global health authorities in the US, EU, and Japan underscores the significant unmet medical need in MSA and the recognition of Amlenetug's potential to address this debilitating condition. These designations provide Lundbeck with substantial incentives and support mechanisms to accelerate the development and potential future review of Amlenetug. The timing of the Fast Track Designation, awarded after the Phase 2 AMULET trial results became available [9], suggests that regulatory authorities, like the FDA, found the overall data package, including the positive signals in the less-impaired patient subgroup [12], sufficiently compelling to warrant expedited development, despite the trial not meeting its primary endpoint in the entire study population. This highlights a pragmatic regulatory approach in the face of severe rare diseases with no existing treatments.

7. Scientific Dissemination

The clinical development and findings related to Amlenetug (Lu AF82422) have been disseminated through peer-reviewed publications and presentations at major international scientific conferences.

Key Publications

A significant publication detailing the Phase 1 study (NCT03611569) is:

  • Buur L, Wiedemann J, Larsen F, et al. Randomized Phase I Trial of the α-Synuclein Antibody Lu AF82422. Mov Disord. 2024;39(6):936-944.[17] This paper reported on the safety, tolerability, pharmacokinetics, and target engagement of ascending single intravenous doses of Lu AF82422 in healthy participants and patients with Parkinson's disease. Key findings included good safety and tolerability, dose-proportional pharmacokinetics, a plasma half-life of approximately 700 hours, and evidence of peripheral and central target engagement through the lowering of free α-synuclein levels in plasma and CSF.[7]

Major Conference Presentations

Data from the Amlenetug clinical program, particularly the Phase 2 AMULET trial, have been presented at prominent international congresses focused on neurology and movement disorders:

  • International Conference on Alzheimer's and Parkinson's Diseases and related neurological disorders (AD/PD) (Lisbon, Portugal, March 2024): The outcomes of the AMULET Phase 2 trial were presented here, forming part of the basis for the FDA's Fast Track Designation.[9]
  • International Congress of Parkinson's Disease and Movement Disorders (MDS) (Philadelphia, PA, September 27-October 1, 2024): Results from the AMULET trial were presented, including an abstract by Singer W, et al., titled "Safety and Efficacy of the Anti-alpha Synuclein Monoclonal Antibody Lu AF82422 for the Treatment of Patients with MSA: Results from the Phase 2 AMULET Trial." (Abstract 20).[12] This presentation detailed the primary endpoint results, subgroup analyses showing more pronounced effects in less impaired patients, and the overall safety profile.
  • International MSA Congress (Boston, U.S., May 9-11, 2025): Lundbeck announced plans to present key pipeline data, patient perspectives from the AMULET trial, and new insights from the MSA natural history study, TALISMAN.[1] Specific presentations include:
  • Poster 8: "Incorporating patient and care partner feedback on the protocol for a clinical trial assessing progression in MSA" (also an oral presentation).
  • Poster 10: "The natural history of multiple system atrophy: a prospective Chinese cohort study."
  • Poster 13: "Amlenetug for the treatment of patients with MSA: A randomized, controlled phase 2 trial" (also an oral presentation by Wolfgang Singer, Mayo Clinic Rochester).

The selection of these high-impact scientific venues for disseminating Amlenetug's data indicates a strategy to engage with specialists and key opinion leaders in the fields of movement disorders and neurodegenerative diseases. This facilitates peer review, critical discussion, and helps to build awareness and scientific consensus regarding the drug's development program and its potential clinical utility.

8. Discussion and Future Outlook

The development of Amlenetug for Multiple System Atrophy represents a significant effort to address a severe unmet medical need. The journey thus far, from preclinical rationale to the initiation of a Phase 3 trial, provides several points for discussion regarding its potential and the challenges ahead.

The efficacy data from the Phase 2 AMULET trial present a nuanced picture. While the trial did not meet its primary endpoint of significantly slowing disease progression in the overall MSA population, the observed 19% slowing with Amlenetug compared to placebo, and particularly the more substantial 37% slowing in the pre-specified subgroup of less-impaired patients, offers a signal of potential biological activity.[12] This finding in the less-impaired subgroup is pivotal, suggesting a potential therapeutic window where intervention earlier in the disease course—before extensive and irreversible neuronal damage has occurred—might be more effective. This hypothesis is common in neurodegenerative disease research, where therapies targeting upstream pathological processes, such as α-synuclein aggregation and spread, are logically expected to yield greater benefits if applied before the downstream neurodegenerative cascade is too advanced. The design of the ongoing Phase 3 MASCOT trial, particularly its patient selection criteria which appear to align with this less-impaired profile (e.g., UMSARS Part I ≤16 [16]), will be critical in formally testing this hypothesis. If confirmed, it would have profound implications not only for the potential clinical use of Amlenetug but also for emphasizing the need for earlier diagnosis and intervention in MSA. The safety profile of Amlenetug observed in Phase 1 and Phase 2 studies has been generally well-tolerated, with no major safety signals precluding further development.[7] The high rate of continuation into the open-label extension of the AMULET trial further supports this.[13]

Targeting α-synuclein remains a cornerstone strategy in the quest for disease-modifying treatments for MSA and other synucleinopathies.[4] Amlenetug's mechanism, aimed at preventing uptake, inhibiting aggregation seeding, and potentially promoting clearance of pathological α-synuclein, is well-grounded in the current understanding of MSA pathogenesis. The collaboration between Lundbeck and Genmab, a company with recognized expertise in antibody technology and development [32], also lends strength to the molecular design and engineering aspects of Amlenetug.[1]

Despite these promising aspects, significant challenges remain. MSA is a clinically heterogeneous disorder, and demonstrating clear disease modification in slowly progressive neurodegenerative conditions is inherently difficult. The robustness and clinical meaningfulness of the Phase 3 MASCOT trial results will be paramount. The trial's global nature and inclusion of different MSA subtypes will provide valuable data, but also add complexity to the analysis.[3]

The primary anticipated milestone for Amlenetug is the outcome of the MASCOT Phase 3 trial. Successful results from this trial could position Amlenetug as a first-in-class disease-modifying therapy for MSA, a significant breakthrough for patients. Beyond MSA, if the underlying principle of targeting extracellular α-synuclein proves effective, it could have broader implications for the development of therapies for other synucleinopathies, although Lundbeck's current focus for Amlenetug appears to be squarely on MSA. The strong regulatory support received through Orphan Drug, Fast Track, and SAKIGAKE designations provides a favorable pathway for Amlenetug's continued development and potential review.[5]

9. Conclusion

Amlenetug (Lu AF82422) stands as a significant investigational candidate in the challenging landscape of Multiple System Atrophy therapeutics. As a human monoclonal antibody targeting extracellular α-synuclein, its mechanism aligns with current understanding of MSA pathogenesis, aiming to prevent the propagation of pathological protein aggregates and potentially slow neurodegeneration.

The clinical development program, while encountering the common hurdle of not meeting its primary endpoint in the overall Phase 2 AMULET trial population, has yielded encouraging signals, particularly in patients with less advanced disease. This observation, combined with a generally favorable safety and tolerability profile, has supported the progression to a large-scale Phase 3 trial (MASCOT). The extensive regulatory support, including Orphan Drug status in major global regions and Fast Track designation in the US, highlights the profound unmet medical need in MSA and the perceived potential of Amlenetug.

The ongoing MASCOT trial will be crucial in determining whether Amlenetug can deliver a clinically meaningful benefit to patients with MSA. Its success would not only offer a much-needed therapeutic option for this devastating rare disease but also provide important validation for α-synuclein-targeted immunotherapies in the broader field of neurodegenerative disorders. The journey of Amlenetug underscores the complexities and perseverance required in developing treatments for rare and progressive neurological conditions.

Works cited

  1. Lundbeck to share pipeline data and key insights into the ..., accessed May 14, 2025, https://www.lundbeck.com/us/newsroom/2025/lundbeck-to-share-pipeline-data-and-key-insights-into-the-progre
  2. Lundbeck to share pipeline data and key insights into the progression of rare disease, Multiple System Atrophy, at International MSA Congress in Boston - PR Newswire, accessed May 14, 2025, https://www.prnewswire.com/news-releases/lundbeck-to-share-pipeline-data-and-key-insights-into-the-progression-of-rare-disease-multiple-system-atrophy-at-international-msa-congress-in-boston-302449751.html
  3. Lundbeck to share pipeline data and key insights into the progression of rare disease, Multiple System Atrophy, at International MSA Congress in Boston - FirstWord Pharma, accessed May 14, 2025, https://firstwordpharma.com/story/5958282
  4. www.lundbeck.com, accessed May 14, 2025, https://www.lundbeck.com/content/dam/lundbeck-com/masters/global-site/global-site/investors/2024/amulet-update/Lu%20AF82422%20phase%20II%20PoC%20read-out%20(AMULET)_31012024.pdf
  5. Multiple System Atrophy Market Size, Share, Trends, Key Companies - DelveInsight, accessed May 14, 2025, https://www.delveinsight.com/report-store/multiple-system-atrophy-msa-market
  6. Lundbeck's amlenetug receives orphan drug status in Japan for MSA, accessed May 14, 2025, https://synapse.patsnap.com/article/lundbecks-amlenetug-receives-orphan-drug-status-in-japan-for-msa
  7. Amlenetug - Drug Targets, Indications, Patents - Patsnap Synapse, accessed May 14, 2025, https://synapse.patsnap.com/drug/9c2178f878a64b83a5b99d47ce4e9d1a
  8. Lundbeck's potential treatment for Multiple System Atrophy granted Orphan Drug Designation in Japan - PR Newswire, accessed May 14, 2025, https://www.prnewswire.com/news-releases/lundbecks-potential-treatment-for-multiple-system-atrophy-granted-orphan-drug-designation-in-japan-302396851.html
  9. Lundbeck's potential treatment amlenetug for Multiple System ..., accessed May 14, 2025, https://www.pharmafocuseurope.com/pharma-exclusives/press-releases/lundbecks-potential-treatment-amlenetug-for-multiple-system-atrophy-receives
  10. Amlenetug - Lundbeck A/S - AdisInsight, accessed May 14, 2025, https://adisinsight.springer.com/drugs/800039097
  11. Lundbeck's potential treatment amlenetug for Multiple System Atrophy receives Fast Track Designation from the FDA, accessed May 14, 2025, https://www.lundbeck.com/us/newsroom/2025/lundbecks-potential-treatment-amlenetug-for-multiple-system-atrophy-fast-track
  12. isctm.org, accessed May 14, 2025, https://isctm.org/public_access/21st_Annual/Poster/Wiedemann-Poster.pdf
  13. FDA Grants Fast Track Designation to Potential Treatment for Multiple System Atrophy, accessed May 14, 2025, https://practicalneurology.com/news/fda-grants-fast-track-designation-to-potential-treatment-for-multisystem-atrophy
  14. FDA Grants Fast Track Designation to Potential Treatment for Multiple System Atrophy, accessed May 14, 2025, https://practicalneurology.com/news/fda-grants-fast-track-designation-to-potential-treatment-for-multisystem-atrophy/2473790/
  15. Safety and Efficacy of the Anti-alpha Synuclein Monoclonal Antibody ..., accessed May 14, 2025, https://www.mdsabstracts.org/abstract/safety-and-efficacy-of-the-anti-alpha-synuclein-monoclonal-antibody-lu-af82422-for-the-treatment-of-patients-with-msa-results-from-the-phase-2-amulet-trial/
  16. FDA Grants Fast Track Designation to Promising Multiple System Atrophy Agent Amlenetug, accessed May 14, 2025, https://www.neurologylive.com/view/fda-grants-fast-track-designation-promising-multiple-system-atrophy-agent-amlenetug
  17. FDA Grants Fast Track Designation to Amlenetug As Treatment for ..., accessed May 14, 2025, https://www.drugtopics.com/view/fda-grants-fast-track-designation-to-amlenetug-as-treatment-for-multiple-system-atrophy
  18. FDA Grants Fast Track Designation to Potential Treatment for Multisystem Atrophy, accessed May 14, 2025, https://practicalneurology.com/news/fda-grants-fast-track-designation-to-potential-treatment-for-multisystem-atrophy?c4src=news:feed
  19. The US government does not review or approve the safety and science of all studies listed on this website. - Error | ClinicalTrials.gov, accessed May 14, 2025, https://clinicaltrials.gov/search?term=NCT06706622&rank=1
  20. 34 - YAbS - The Antibody Society, accessed May 14, 2025, https://db.antibodysociety.org/db0/results_target?sort=dt_phase1&page=34
  21. Amlenetug Biosimilar - Anti-Alpha-synuclein mAb - Research Grade - ProteoGenix US, accessed May 14, 2025, https://us.proteogenix.science/product/amlenetug-biosimilar-anti-alpha-synuclein-mab-research-grade/
  22. Emerging targets of α-synuclein spreading in α-synucleinopathies: a ..., accessed May 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11756073/
  23. An update on immune-based alpha-synuclein trials in Parkinson's ..., accessed May 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11638298/
  24. Disease-Modifying Therapies for Multiple System Atrophy: Where Are We in 2022? - PMC, accessed May 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9398078/
  25. H. Lundbeck, accessed May 14, 2025, https://www.lundbeck.com/content/dam/lundbeck-com/masters/global-site/global-site/investors/reports-and-presentations-/2025/jpm-2025/Webcast%20transcript%20JPM%202025%20-%20H.%20Lundbeck.pdf
  26. Disease-Modifying Therapies for Multiple System Atrophy: Where Are We in 2022?, accessed May 14, 2025, https://www.researchgate.net/publication/360343700_Disease-Modifying_Therapies_for_Multiple_System_Atrophy_Where_Are_We_in_2022
  27. Trial of LU AF82422 in participants with multiple system atrophy (H-56562), accessed May 14, 2025, https://cdn.bcm.edu/healthcare/clinical-trials/h-56562
  28. Amlenetug Recombinant Antibody - SNCA - MyBioSource, accessed May 14, 2025, https://www.mybiosource.com/human-recombinant-antibody/amlenetug/1581355
  29. Randomized Phase I Trial of the α-Synuclein Antibody Lu AF82422, accessed May 14, 2025, https://pubmed.ncbi.nlm.nih.gov/38494847/
  30. Analysis of novel immunotherapy approaches in the treatment of Parkinson's Disease, accessed May 14, 2025, https://www.researchgate.net/publication/384435546_Analysis_of_novel_immunotherapy_approaches_in_the_treatment_of_Parkinson's_Disease
  31. A-Synuclein Antibody Lu AF82422 Shows Disease-Modifying Potential in Phase 2 AMULET Trial of MSA - NeurologyLive, accessed May 14, 2025, https://www.neurologylive.com/view/a-synuclein-antibody-lu-af82422-disease-modifying-potential-phase-2-amulet-trial-msa
  32. Antibody Science and Innovation - Genmab, accessed May 14, 2025, https://www.genmab.com/antibody-science
  33. A Unique Approach to Advancing Antibody Science - Genmab US, accessed May 14, 2025, https://www.us.genmab.com/research/our-unique-approach

Published at: May 14, 2025

This report is continuously updated as new research emerges.

© Copyright 2025. All Rights Reserved by MedPath