MedPath

Prasinezumab Advanced Drug Monograph

Published:May 8, 2025

Generic Name

Prasinezumab

Drug Type

Biotech

CAS Number

1960462-19-4

Prasinezumab (DB14788): An Investigational Anti-Alpha-Synuclein Antibody for Parkinson's Disease

1. Executive Summary

Prasinezumab (DB14788) is an investigational humanized monoclonal antibody targeting aggregated alpha-synuclein (α-syn), a protein central to the pathology of Parkinson's disease (PD). Developed collaboratively by Prothena and Roche, Prasinezumab aims to slow disease progression by inhibiting the accumulation and spread of pathogenic α-synuclein aggregates. Clinical development has progressed through Phase 2b, evaluating the therapy primarily in early-stage PD populations.

Key findings indicate that Prasinezumab possesses a generally favorable safety and tolerability profile, demonstrated across multiple clinical trials, including long-term open-label extensions involving over 900 participants. Immunogenicity concerns have not been significant. However, efficacy results have been mixed. While Phase 1 studies showed peripheral target engagement, the pivotal Phase 2 PASADENA (Part 1) and Phase 2b PADOVA trials failed to meet their primary endpoints assessing overall clinical progression or time to motor progression, respectively, in the intent-to-treat populations over 52-104 weeks. Despite these outcomes, consistent signals suggesting a slowing of motor decline, specifically measured by the Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) Part III, were observed in both trials, particularly in pre-specified subgroup analyses (e.g., patients on levodopa in PADOVA) and in long-term (4-year) open-label extension data compared to external controls.

Potential limitations to efficacy include the antibody's inability to bind C-terminally truncated α-synuclein species found in PD brains and challenges in translating preclinical findings from animal models. Roche is currently evaluating the comprehensive data from the PADOVA trial and ongoing open-label extensions to determine the future development pathway and regulatory strategy for Prasinezumab, with potential filings projected for 2027 or later. Prasinezumab represents a significant effort in targeting α-synuclein pathology, but definitive proof of its disease-modifying efficacy in controlled trials remains elusive.

2. Introduction

2.1. Parkinson's Disease Pathophysiology

Parkinson's disease (PD) is a progressive neurodegenerative disorder primarily characterized by motor symptoms such as bradykinesia, rigidity, resting tremor, and postural instability, alongside a spectrum of non-motor symptoms including cognitive impairment, sleep disturbances, and autonomic dysfunction.[1] Pathologically, PD is defined by the loss of dopaminergic neurons, particularly in the substantia nigra pars compacta, and the presence of intraneuronal proteinaceous inclusions known as Lewy bodies and Lewy neurites.[1] A principal component of these inclusions is misfolded and aggregated alpha-synuclein (α-syn).[1] The accumulation and potential cell-to-cell propagation of these α-syn aggregates are believed to play a central role in the neurodegenerative process underlying PD.[2]

2.2. Unmet Need for Disease-Modifying Therapies (DMTs)

Current therapeutic strategies for PD, such as levodopa and dopamine agonists, primarily provide symptomatic relief by compensating for dopamine deficiency.[2] While effective in managing motor symptoms, particularly in the early stages, these treatments do not address the underlying neurodegenerative process or prevent the progressive loss of neurons.[2] As the disease advances, the efficacy of symptomatic treatments often wanes, and motor complications like dyskinesia and 'on-off' fluctuations frequently emerge.[2] Consequently, there is a critical unmet medical need for disease-modifying therapies (DMTs) capable of slowing, halting, or reversing the progression of PD.[4] The development of such therapies represents a major focus of current PD research.[7]

2.3. Immunotherapy Targeting Alpha-Synuclein

Given the central role of α-syn aggregation in PD pathology, therapeutic strategies aimed at clearing or preventing the spread of these pathological protein forms are under active investigation.[4] Immunotherapy, involving either active vaccination to stimulate the patient's immune system against α-syn or passive administration of monoclonal antibodies targeting α-syn, represents a prominent approach.[2] The rationale is that antibodies can neutralize extracellular α-syn aggregates, preventing their uptake by neighboring neurons and thus inhibiting propagation, and potentially facilitate clearance via mechanisms like microglial phagocytosis or lysosomal pathways.[4] Despite strong preclinical evidence supporting this strategy, translating these approaches into clinically successful DMTs has proven challenging, with several candidates facing setbacks in clinical trials.[2]

2.4. Prasinezumab Overview

Prasinezumab (formerly known as PRX002 or RG7935) is an investigational humanized IgG1 monoclonal antibody specifically designed to target aggregated forms of α-synuclein.[1] Developed initially by Prothena Biosciences and subsequently licensed to Roche for global development and commercialization, Prasinezumab represents a passive immunotherapy approach aimed at slowing PD progression.[1] It is administered intravenously and has been evaluated in clinical trials focusing on patients with early-stage Parkinson's disease.[1]

3. Prasinezumab: Profile and Mechanism of Action

3.1. Drug Identification and Characteristics

Prasinezumab is a biological entity, specifically a humanized IgG1 monoclonal antibody.[13] It originated from the murine monoclonal antibody 9E4.[8] Key identifiers are summarized in Table 1.

Table 1: Key Identifiers for Prasinezumab

IdentifierValueSource(s)
NamePrasinezumab12
Alternative NamesPRX002, RG79351
DrugBank IDDB14788(User Query)
TypeBiotech, Monoclonal Antibody (Humanized IgG1)13
CAS Number1960462-19-413
Originator/DeveloperProthena / Roche3

3.2. Target and Binding Specificity

Prasinezumab is engineered to selectively target and bind to aggregated forms of α-synuclein, which are considered pathogenic in PD.[4] Its binding site is a specific epitope located within the C-terminus of the human α-synuclein protein, corresponding to amino acids 118–126.[1] Preclinical characterization indicates that Prasinezumab binds to human aggregated α-synuclein with high affinity and avidity.[13]

3.3. Proposed Therapeutic Mechanism

The intended therapeutic mechanism of Prasinezumab is centered on mitigating the neurotoxicity associated with α-synuclein aggregates.[4] By binding to these aggregates, the antibody aims to achieve several downstream effects:

  1. Inhibition of Accumulation: Preventing the further build-up of toxic α-syn species within the brain.[4]
  2. Inhibition of Spreading: Blocking the cell-to-cell transmission of pathogenic α-synuclein conformers, a process thought to drive disease progression.[1]
  3. Facilitation of Clearance: Potentially engaging clearance mechanisms, such as the lysosomal pathway, to remove existing aggregates.[11]

Through these actions, Prasinezumab is hypothesized to protect neurons from α-synuclein-mediated damage and ultimately slow the clinical progression of Parkinson's disease.[4]

3.4. Preclinical Rationale and Potential Limitations

The development of Prasinezumab is supported by a substantial body of preclinical evidence implicating α-synuclein aggregation in PD pathogenesis.[4] Studies utilizing the parent mouse antibody (9E4) in animal models of PD demonstrated promising results, including reduced α-synuclein accumulation in the brain, protection of dopaminergic neurons, decreased cell-to-cell propagation of pathology, and improvements in motor and cognitive deficits.[1]

However, certain aspects of Prasinezumab's binding characteristics and the models used raise potential limitations:

  • C-Terminally Truncated α-Synuclein: A significant consideration is the existence of C-terminally truncated forms of α-synuclein (e.g., species ending at amino acid 103, 110, or 115) identified in post-mortem PD brain tissue.[8] These truncated forms exhibit a high propensity for aggregation and are considered pathogenic.[8] Because Prasinezumab's epitope lies within the C-terminus (amino acids 118-126), it is unable to recognize or bind these truncated species.[8] This inability to target a potentially important pool of toxic, aggregation-prone α-synuclein could limit the overall therapeutic efficacy of the antibody in clearing the full spectrum of relevant pathology in the human brain. This disconnect might contribute to the observed discrepancies between preclinical promise and clinical outcomes.
  • Translatability of Animal Models: While mouse models provided proof-of-concept, inherent differences exist between human and mouse α-synuclein sequences and processing.[8] Human α-synuclein seeds propagate aggregation more slowly in mice and vice-versa.[8] These species-specific differences mean that mouse models may not fully recapitulate human PD pathology or accurately predict the efficacy of therapies targeting human α-synuclein.[8] This limitation underscores the challenge in translating findings from preclinical models to human clinical trials for α-synuclein-targeted therapies.

4. Clinical Development and Efficacy

4.1. Overview of Clinical Program

Prasinezumab's clinical development has primarily focused on evaluating its potential as a DMT in individuals with early-stage Parkinson's disease.[1] The program, led by Roche in collaboration with Prothena, has progressed through Phase 1 safety and tolerability studies, a Phase 2 efficacy study (PASADENA), and a subsequent Phase 2b study (PADOVA) designed to further investigate signals observed in Phase 2.[1] Open-label extension (OLE) phases for both PASADENA and PADOVA are ongoing to assess long-term effects.[3] Table 2 summarizes the key clinical trials.

Table 2: Summary of Key Prasinezumab Clinical Trials

Trial IDPhaseTitle/PurposeKey PopulationPrimary Endpoint(s)Key Findings Summary
NCT02095171Phase 1Safety, Tolerability, PK/PD of single ascending dosesHealthy Volunteers (n=40)Safety & TolerabilitySafe & well-tolerated up to 30 mg/kg. Dose-dependent reduction in serum free α-synuclein.1
NCT02157714Phase 1bSafety, Tolerability, PK/PD of multiple ascending dosesMild-Moderate PD (n=80)Safety & TolerabilityGenerally safe & well-tolerated (0.3-60 mg/kg x3 q4w). 2 discontinuations (infusion reaction) at highest dose. Serum α-syn reduction. No significant clinical (MDS-UPDRS) or CSF α-syn change.1
NCT03100149Phase 2PASADENA: Efficacy & Safety of Prasinezumab in Early PD (Part 1: 52 wks; Part 2: 52 wks OLE)Early PD (≤2 yrs diagnosis, H&Y I-II), drug-naïve or MAO-Bi (n=316)Part 1: Change in MDS-UPDRS Total Score (I+II+III) at Wk 52Part 1: Missed primary endpoint. Signal of slowed motor progression (MDS-UPDRS Part III). No difference on DaT-SPECT. Favorable safety/tolerability.14 Post-hoc: Greater effect in faster progressors.22 OLE (4 yrs vs. PPMI): Sustained slowing of motor progression (MDS-UPDRS III OFF/ON) & ADLs (MDS-UPDRS II).12
NCT04777331Phase 2bPADOVA: Efficacy & Safety of Prasinezumab in Early PD on Stable Symptomatic Tx (min. 18 mos; ongoing OLE)Early PD (H&Y 1-2) on stable Levodopa or MAO-Bi (n=586)Time to Confirmed Motor Progression (≥5 pt increase MDS-UPDRS Part III OFF state)Missed primary endpoint (overall population: p=0.0657). Signal stronger in Levodopa subgroup (75% pts, nominal p=0.0431). Positive trends on secondary/exploratory endpoints. Well-tolerated, no new safety signals.12

(PD: Parkinson's Disease; PK/PD: Pharmacokinetics/Pharmacodynamics; α-syn: alpha-synuclein; MDS-UPDRS: Movement Disorder Society-Unified Parkinson's Disease Rating Scale; H&Y: Hoehn & Yahr Stage; MAO-Bi: Monoamine Oxidase B inhibitor; DaT-SPECT: Dopamine Transporter Single Photon Emission Computed Tomography; OLE: Open-Label Extension; PPMI: Parkinson's Progression Markers Initiative; ADL: Activities of Daily Living; Tx: Treatment; HR: Hazard Ratio; CI: Confidence Interval; CSF: Cerebrospinal Fluid; pts: patients)

4.2. Phase 1 Studies

Initial Phase 1 studies established the preliminary safety profile and pharmacokinetic/pharmacodynamic properties of Prasinezumab. A single ascending dose study (NCT02095171) in 40 healthy volunteers found single intravenous infusions up to 30 mg/kg to be safe and well-tolerated.[1] Importantly, it demonstrated peripheral target engagement, with dose-dependent reductions in free serum α-synuclein levels observed shortly after infusion, reaching up to 97% reduction at the highest dose.[1]

A subsequent Phase 1b multiple ascending dose study (NCT02157714) involved 80 participants with mild-to-moderate PD receiving three intravenous infusions every four weeks at doses up to 60 mg/kg.[1] The drug was again generally well-tolerated, although two participants discontinued due to infusion-related reactions at the 60 mg/kg dose.[1] Similar reductions in serum α-synuclein were observed.[1] However, this study provided an early indication of the challenges ahead: no significant clinical improvement based on MDS-UPDRS scores was detected, nor were there changes in CSF α-synuclein levels, suggesting a potential disconnect between peripheral target engagement and central nervous system effects or clinical benefit at this stage.[9]

4.3. Phase 2 PASADENA Trial (NCT03100149)

4.3.1. Design and Population

The PASADENA study was a large, multicenter, randomized, double-blind, placebo-controlled Phase 2 trial designed to evaluate the efficacy and safety of Prasinezumab over a longer duration in early PD.[5] Part 1 involved 316 participants randomized (1:1:1) to receive intravenous infusions of low-dose (1500 mg), high-dose (3500 mg for <65 kg, 4500 mg for ≥65 kg), or placebo every 4 weeks for 52 weeks.[14] Eligible participants had been diagnosed with PD within the previous two years, were Hoehn & Yahr (H&Y) stage I or II, and were either treatment-naïve or on stable MAO-B inhibitor therapy only.[5] Baseline characteristics showed a mean age of ~60 years and mean time since diagnosis of ~10 months.[1] Part 2 was a 52-week blinded OLE where all participants received active treatment.[15]

4.3.2. Endpoints

The primary endpoint for Part 1 was the change from baseline (CFB) to Week 52 in the MDS-UPDRS total score (sum of Parts I, II, and III) compared to placebo.[14] Key secondary endpoints included CFB in DaT-SPECT signal in the putamen, CFB in MDS-UPDRS Part III (motor score), Clinical Global Impression of Improvement (CGI-I), Patient Global Impression of Change (PGIC), and time to initiation of symptomatic dopaminergic treatment.[14]

4.3.3. PASADENA Part 1 Results (52 Weeks)

The PASADENA trial did not meet its primary endpoint at 52 weeks.[14] The change in MDS-UPDRS total score was not significantly different between either Prasinezumab dose group and the placebo group (Difference vs placebo: Low dose -2.02 points, 80% CI -4.21 to 0.18; High dose -0.62 points, 80% CI -2.82 to 1.58).[14] Similarly, there was no significant difference in the progression of dopamine transporter deficit as measured by DaT-SPECT imaging, although overall progression on this measure was limited (<11%) in all groups over the 52 weeks.[14]

Despite missing the primary endpoint, a signal of efficacy was observed on motor progression, specifically the change in MDS-UPDRS Part III score.[14] Participants treated with Prasinezumab showed less worsening on this motor scale compared to placebo (Difference vs placebo: Low dose -1.88 points; High dose -1.02 points).[14] This finding was supported by exploratory analyses, including centrally rated MDS-UPDRS Part III scores and digital biomarker assessments.[14]

4.3.4. Post-Hoc Subgroup Analysis

Further exploratory post-hoc analyses suggested that the potential benefit of Prasinezumab on slowing motor progression (MDS-UPDRS Part III) might be more pronounced in certain subgroups characterized by faster disease progression.[8] These subgroups included participants with a "diffuse malignant" phenotype or those who were already taking MAO-B inhibitors at baseline.[22] While such post-hoc analyses must be interpreted with caution due to the increased risk of false positives and potential confounding factors [8], they generated hypotheses about patient heterogeneity influencing treatment response. The observation that a signal was detected specifically on the motor subscale (Part III), but not the composite primary endpoint, highlights the complexities in selecting appropriate endpoints for PD DMT trials, especially over relatively short durations like 52 weeks. These findings significantly influenced the design and focus of the subsequent PADOVA trial.

4.3.5. Open-Label Extension (OLE) Results (4 Years)

Long-term follow-up data from the PASADENA OLE provided further evidence supporting a potential effect on motor progression.[12] This analysis compared participants who received Prasinezumab from the start ("early-start" group, n=177) and those who switched from placebo to Prasinezumab after 52 weeks ("delayed-start" group, n=94) against an external comparator cohort derived from the Parkinson's Progression Markers Initiative (PPMI) observational study (n=303) over 4 years.[23]

The results indicated that both the early-start and delayed-start Prasinezumab groups exhibited significantly slower motor progression, as measured by MDS-UPDRS Part III scores in both the OFF and ON medication states, compared to the PPMI cohort.[12] The relative slowing of decline on MDS-UPDRS Part III OFF state was estimated at approximately 51% for the delayed-start group and 65% for the early-start group compared to PPMI.[12] Similar sustained benefits were observed for patient-reported activities of daily living (MDS-UPDRS Part II).[23]

While comparisons to external control groups have inherent limitations (e.g., potential differences in populations, data collection methods), these 4-year OLE findings offer the most compelling suggestion to date of a sustained, long-term disease-modifying effect of Prasinezumab on motor aspects of PD. The contrast between these long-term observations and the lack of significant effect in the initial 52-week controlled phase underscores the potential need for extended trial durations to detect DMT effects in slowly progressing neurodegenerative diseases like PD, or suggests a possible delayed onset or cumulative benefit of the therapy.

4.4. Phase 2b PADOVA Trial (NCT04777331)

4.4.1. Rationale and Design

Based on the motor signal observed in PASADENA, the PADOVA trial was designed specifically to confirm this effect, using a time-to-event primary endpoint focused on motor progression.[16] It was a Phase 2b, multicenter, randomized, double-blind, placebo-controlled study evaluating Prasinezumab 1500 mg administered intravenously every 4 weeks versus placebo.[16] A key difference from PASADENA was the inclusion of participants already on stable standard-of-care (SOC) symptomatic PD medication (levodopa or MAO-B inhibitor monotherapy).[16] The trial enrolled 586 participants treated for a minimum of 18 months.[12]

4.4.2. Population

The PADOVA population consisted of individuals with early-stage PD (H&Y 1-2).[16] Compared to the PASADENA cohort, PADOVA participants were, on average, about 5 years older, had a longer disease duration by approximately 8 months, and exhibited slightly more advanced disease severity at baseline based on H&Y stage and MDS-UPDRS Part III scores.[16] At baseline, 74.2% were receiving levodopa and 25.8% were receiving an MAO-B inhibitor.[16] Detailed inclusion and exclusion criteria can be found in sources.[31]

4.4.3. Primary Endpoint Results

The PADOVA trial failed to meet its primary endpoint of time to confirmed motor progression (defined as ≥5 point increase in MDS-UPDRS Part III OFF state) with statistical significance in the overall study population.[4] The hazard ratio (HR) for progression was 0.84 (95% CI: 0.69-1.01), with a p-value of 0.0657.[4]

4.4.4. Subgroup and Secondary Findings

Consistent with the hypothesis-generating findings from PASADENA, a pre-specified analysis of the subgroup of participants treated with levodopa (representing 75% of the trial population) showed a more pronounced effect.[1] In this subgroup, Prasinezumab was associated with a nominally statistically significant reduction in the risk of motor progression (HR=0.79, 95% CI: 0.63-0.99, nominal p=0.0431).[4] Furthermore, pre-specified supplementary covariate-adjusted analyses yielded nominally significant results for both the overall population (HR=0.81, nominal p=0.0334) and the levodopa subgroup (HR=0.76, nominal p=0.0175).[12]

Roche also reported consistent positive trends favoring Prasinezumab across multiple secondary and exploratory endpoints, including measures of motor function, global impression of change, and digital health tool assessments, although specific data were not provided in the reviewed sources.[4]

The PADOVA results mirrored the pattern seen in PASADENA: failure to achieve the primary endpoint in the overall population, but persistent signals suggestive of a benefit on motor progression, particularly evident in a large, pre-specified subgroup (levodopa users). The reason for the stronger signal in levodopa users remains unclear and could relate to specific patient characteristics within that group, a potential interaction with levodopa, or statistical chance. This finding, while not conclusive due to the lack of overall primary endpoint significance, is a key factor in ongoing evaluations by Roche.

5. Safety, Tolerability, and Immunogenicity

5.1. Integrated Safety Summary

Across the clinical development program, including Phase 1, Phase 2 (PASADENA), Phase 2b (PADOVA), and long-term OLEs, Prasinezumab has demonstrated a generally favorable safety and tolerability profile.[1] The safety database encompasses over 900 PD participants, with more than 500 individuals having received treatment for periods ranging from 1.5 to 5 years.[4] Importantly, the PADOVA trial did not identify any new safety signals.[4] Key safety findings are summarized in Table 3.

Table 3: Summary of Key Safety Findings for Prasinezumab

Trial Phase/NameKey Tolerability FindingsCommon Adverse Events (AEs)Serious Adverse Events (SAEs)Immunogenicity Notes
Phase 1 (Healthy/PD)Generally well-tolerated up to highest doses tested (30 mg/kg single; 60 mg/kg multiple).1Infusion-related reactions (led to 2 discontinuations at 60 mg/kg in Phase 1b).1Not specified in detail.Not specified.
Phase 2 PASADENA (Part 1)Generally well tolerated, favorable safety profile.14Infusion reactions (19-34%), nasopharyngitis, back pain, headache.1No life-threatening AEs reported.14 Mention of suicidal events requires monitoring.8No immunogenicity concerns reported.14 ADA assessed as secondary endpoint.26
Phase 2b PADOVAWell tolerated.4Not specified in detail, but consistent with previous trials.12No new safety signals observed.4Not specified.
Overall (>900 pts, up to 5 yrs)Generally well tolerated in long-term exposure.4Infusion-related reactions most notable.1Not specified in detail.Low immunogenicity expected for humanized IgG1.34

(PD: Parkinson's Disease; AE: Adverse Event; SAE: Serious Adverse Event; ADA: Anti-Drug Antibody; pts: patients)

5.2. Common and Serious Adverse Events

The most frequently reported adverse events associated with Prasinezumab treatment across trials have been infusion-related reactions (such as rash or nausea), nasopharyngitis (common cold symptoms), back pain, and headache.[1] Infusion reactions occurred in 19% to 34% of participants in Phase 2 [27] and led to two discontinuations at the highest dose tested (60 mg/kg) in the Phase 1b study.[1] No life-threatening adverse events were reported in the 52-week PASADENA Part 1 analysis.[14] While a review noted the occurrence of suicidal events in Prasinezumab-treated groups, causality was not established, but close monitoring was recommended.[8]

5.3. Immunogenicity Assessment

The development of anti-drug antibodies (ADAs) can impact the efficacy and safety of biologic therapies. Immunogenicity was assessed as a secondary endpoint in the PASADENA trial.[26] Results from PASADENA Part 1 indicated no immunogenicity concerns.[14] This aligns with the expectation of low immunogenicity for humanized IgG1 monoclonal antibodies like Prasinezumab.[34]

The consistent reporting of good tolerability and the absence of significant safety or immunogenicity issues, even with long-term administration in the OLEs, represent a positive aspect of Prasinezumab's development profile. This favorable safety data may support continued investigation despite the challenges in demonstrating robust efficacy in the controlled phases of the trials.

6. Regulatory Status and Future Outlook

6.1. Current Development Stage

Prasinezumab is currently in a post-Phase 2b stage of development.[3] The pivotal controlled trials, PASADENA (Phase 2) and PADOVA (Phase 2b), have completed their primary analysis periods.[12] Open-label extension studies for both trials are ongoing to gather longer-term data on safety and efficacy.[3] Development is led by Roche, under a licensing agreement with Prothena.[3]

6.2. Regulatory Interactions and Potential Timelines

Following the PADOVA trial results, Roche has stated its intention to further evaluate the comprehensive dataset and engage with health authorities, such as the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA), to determine the appropriate next steps for the program.[12] Based on available information, a potential regulatory filing for market approval is not anticipated in the near term, with projections suggesting 2027 or beyond.[19]

6.3. Regulatory Designations

Parkinson's disease, while serious, does not typically meet the prevalence criteria for Orphan Drug Designation in major markets.[8] Some Prothena corporate materials mention Fast Track designation by the FDA for Prasinezumab.[34] However, other sources specifically link Fast Track designation to different assets in Prothena's pipeline (Birtamimab, PRX123).[35] Therefore, confirmation of Fast Track status specifically for Prasinezumab requires clarification from definitive regulatory sources.

6.4. Positioning within the PD Therapeutic Landscape

Prasinezumab is one of several immunotherapies targeting α-synuclein currently or recently in clinical development, employing both passive (monoclonal antibodies) and active (vaccines) strategies.[2] It represents a potential DMT approach, aiming to address the underlying pathology, which distinguishes it from existing symptomatic treatments.[2] The field of PD DMTs is evolving rapidly, with other approaches targeting different mechanisms, such as LRRK2 inhibition (e.g., BIIB122), GBA pathway enhancement (e.g., Ambroxol, PR001/LY3884961), GLP-1 receptor activation (e.g., Lixisenatide), and neuroinflammation, also advancing through the pipeline.[6]

The future trajectory for Prasinezumab appears uncertain following the PADOVA results. Roche's non-committal statements regarding next steps and the distant projected filing timeline suggest that the current data package, particularly the missed primary endpoints in controlled settings, may not be sufficient to proceed directly to Phase 3 trials without further supportive evidence.[4] The decision likely awaits deeper analysis of the PADOVA secondary and exploratory endpoints, results from the ongoing OLEs, and discussions with regulatory agencies regarding the path forward. Identifying a specific patient subgroup that clearly benefits or validating a responsive biomarker could be critical for future development.

7. Discussion

The development of Prasinezumab exemplifies both the promise and the challenges inherent in targeting α-synuclein pathology for Parkinson's disease modification. The biological rationale remains strong, supported by extensive preclinical work.[4] However, translating this rationale into unequivocal clinical efficacy has proven difficult, as evidenced by the mixed results from the PASADENA and PADOVA trials.[8]

A key point for discussion is the apparent discrepancy between the primary outcomes of the controlled trial phases and the signals observed on motor subscales and in longer-term or subgroup analyses.[12] The failure to meet primary endpoints (MDS-UPDRS Total Score in PASADENA Part 1; Time to Motor Progression in PADOVA) could stem from several factors. The chosen endpoints might lack sensitivity to detect subtle disease modification over the 1-2 year timeframe of the controlled phases, especially in early PD where progression can be slow and variable.[28] Patient heterogeneity is another critical factor; PD is increasingly recognized as a complex syndrome with diverse underlying drivers, meaning not all patients may benefit equally from an α-synuclein-targeted therapy.[8] The post-hoc findings in PASADENA and the subgroup analysis in PADOVA hinting at greater effects in faster-progressing patients or those on levodopa support this notion.[12] Alternatively, the therapeutic effect might have a delayed onset or require prolonged treatment to become clinically meaningful, as suggested by the positive OLE results.[23]

The limitations inherent in the therapeutic approach itself must also be considered. The inability of Prasinezumab to bind C-terminally truncated α-synuclein, a known pathogenic species, represents a potential gap in target coverage.[8] Furthermore, challenges remain regarding the optimal targeting strategy (intracellular vs. extracellular α-synuclein), antibody penetration into the CNS and specific microenvironments like the synaptic cleft, and the fidelity of preclinical models in predicting human responses.[8]

The lack of robust biomarker changes correlating with clinical signals further complicates interpretation. While peripheral α-synuclein reduction was achieved, CSF levels were unchanged in Phase 1b [9], and DaT-SPECT imaging showed no treatment effect in PASADENA.[14] While exploratory imaging biomarkers in PADOVA (neuromelanin, iron accumulation) showed potentially positive trends [33], the field urgently needs validated biomarkers that reliably track target engagement within the CNS and correlate with disease modification to guide development.[7]

Future directions for Prasinezumab, should development continue, might involve trials specifically designed for enriched patient populations (e.g., based on progression rate, biomarker status, or potentially levodopa use), utilizing sensitive endpoints possibly including digital measures, and ensuring sufficient duration to capture long-term effects. Confirmation of the OLE findings in a rigorously controlled setting would be crucial.

8. Conclusion

Prasinezumab is an investigational monoclonal antibody targeting aggregated α-synuclein, developed as a potential disease-modifying therapy for Parkinson's disease. Clinical trials have established a generally favorable safety and tolerability profile, even with long-term administration. However, efficacy results have been inconsistent. While the Phase 2 PASADENA and Phase 2b PADOVA trials did not meet their primary endpoints in the overall early PD populations studied, they generated signals suggestive of a slowing effect on motor progression (MDS-UPDRS Part III). These signals were more apparent in post-hoc/subgroup analyses of potentially faster-progressing patients and in long-term open-label extension data compared to external controls.

Significant challenges remain, including the need to definitively demonstrate efficacy in robust, controlled trials, understanding the impact of patient heterogeneity, addressing potential limitations in target coverage (e.g., truncated α-synuclein), and identifying reliable biomarkers of target engagement and disease modification. Roche, the lead developer, is currently evaluating the comprehensive data package to determine the future development strategy in consultation with regulatory authorities. Prasinezumab remains an investigational agent whose potential as a disease-modifying therapy for Parkinson's disease requires further validation.

9. References

[1]

Works cited

  1. Prasinezumab for Parkinson's disease, accessed May 8, 2025, https://parkinsonsnewstoday.com/prx002/
  2. The safety and effectiveness of α-synuclein immunotherapy vs. placebo for the treatment of Parkinson's disease: a systematic review and meta-analysis - Annals of Palliative Medicine, accessed May 8, 2025, https://apm.amegroups.org/article/view/107173/html
  3. Parkinson's Disease - Prothena, accessed May 8, 2025, https://www.prothena.com/pipeline/parkinsons-disease/
  4. Roche's Phase IIb study of prasinezumab missed primary endpoint ..., accessed May 8, 2025, https://www.roche.com/media/releases/med-cor-2024-12-19
  5. A Phase II Study to Evaluate the Safety and Efficacy of Prasinezumab in Early Parkinson's Disease (PASADENA): Rationale, Design, and Baseline Data - PMC, accessed May 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8518716/
  6. The Evolving Landscape of Gene and Cell Therapies in Parkinson Disease: Pipeline Highlights - CGTLive®, accessed May 8, 2025, https://www.cgtlive.com/view/evolving-landscape-gene-cell-therapies-parkinson-disease
  7. Disease-Modifying Research Pipeline Holds Possibility for Parkinson's, accessed May 8, 2025, https://www.parkinson.org/blog/research/disease-modifying-research
  8. Prasinezumab slows motor progression in Parkinsons disease: beyond the clinical data, accessed May 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11839931/
  9. An update on immune-based alpha-synuclein trials in Parkinson's ..., accessed May 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11638298/
  10. pmc.ncbi.nlm.nih.gov, accessed May 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11638298/#:~:text=In%20conclusion%2C%20using%20immunotherapy%20to,the%20need%20for%20further%20research.
  11. pmc.ncbi.nlm.nih.gov, accessed May 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8518716/#:~:text=Although%20blockade%20of%20cell%2Dto,the%20lysosomal%20pathway%20(12).
  12. Roche's Phase IIb Study of Prasinezumab Missed Primary Endpoint, But Suggests Possible Clinical Benefit in Early-Stage Parkinson's Disease - Business Wire, accessed May 8, 2025, https://www.businesswire.com/news/home/20241218381145/en/Roches-Phase-IIb-Study-of-Prasinezumab-Missed-Primary-Endpoint-But-Suggests-Possible-Clinical-Benefit-in-Early-Stage-Parkinsons-Disease
  13. Prasinezumab | CAS NO.:1960462-19-4 - GlpBio, accessed May 8, 2025, https://www.glpbio.com/sp/prasinezumab.html
  14. PASADENA: A Phase 2 study to evaluate the safety and efficacy of prasinezumab in early Parkinson's disease; Part 1 Week-52 results - MDS Abstracts, accessed May 8, 2025, https://www.mdsabstracts.org/abstract/pasadena-a-phase-2-study-to-evaluate-the-safety-and-efficacy-of-prasinezumab-in-early-parkinsons-disease-part-1-week-52-results/
  15. A study to evaluate whether Prasinezumab can slow or halt disease progression in people with a recent diagnosis of Parkinson's - ForPatients, accessed May 8, 2025, https://forpatients.roche.com/bin/medically/download-clinical-trial?id=NCT03100149-2017-000087-152023-504472-24-00-BP39529
  16. A Phase 2b, multicenter, randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of intravenous prasinezumab in early-stage Parkinson's disease (PADOVA): Rationale, design, and baseline data - PubMed, accessed May 8, 2025, https://pubmed.ncbi.nlm.nih.gov/39798255/
  17. 231 A Phase2 study evaluating safety and efficacy of prasinezumab in early Parkinson's disease (PASADENA) | Journal of Neurology, Neurosurgery & Psychiatry, accessed May 8, 2025, https://jnnp.bmj.com/content/93/6/A80.1
  18. Prasinezumab | 1960462-19-4 - ChemicalBook, accessed May 8, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB38081057.htm
  19. Prasinezumab - Prothena Corporation/Roche - AdisInsight, accessed May 8, 2025, https://adisinsight.springer.com/drugs/800040327
  20. A Study to Evaluate the Efficacy and Safety of Intravenous Prasinezumab in Participants with Early Parkinson's Disease (PADOVA): Rationale, Design, and Baseline Data - Roche Science Hub, accessed May 8, 2025, https://medically.roche.com/global/en/neuroscience/mds-2024/medical-material/MDS-2024-poster-nikolcheva-a-study-to-evaluate-the-efficacy-and-safety-pdf.html
  21. Prasinezumab slows motor progression, misses main trial goal, accessed May 8, 2025, https://parkinsonsnewstoday.com/news/prasinezumab-slows-motor-progression-misses-main-trial-goal/
  22. Prasinezumab slows motor progression in rapidly progressing early-stage Parkinson's disease - PubMed, accessed May 8, 2025, https://pubmed.ncbi.nlm.nih.gov/38622249/
  23. Sustained effect of prasinezumab on Parkinson's disease motor ..., accessed May 8, 2025, https://pubmed.ncbi.nlm.nih.gov/39379705/
  24. Effect of Prasinezumab on Parkinson's Disease Motor Progression in a Long-term Open-label Extension of the PASADENA Trial, accessed May 8, 2025, https://medically.roche.com/global/en/neuroscience/mds-2024/medical-material/MDS-2024-poster-pagano-effect-of-prasinezumab-pdf.html
  25. Prasinezumab Falls Short of Statistical Significance in Primary Endpoint of PADOVA Trial for Parkinson Disease, accessed May 8, 2025, https://www.appliedclinicaltrialsonline.com/view/prasinezumab-primary-endpoint-padova-trial-parkinson-disease
  26. A study to evaluate the efficacy of Prasinezumab (RO7046015/PRX002) in participants with early Parkinson's Disease (PASADENA), accessed May 8, 2025, https://www.journalofparkinsonsdisease.com/study-evaluate-efficacy-prasinezumab-ro7046015prx002-participants-early-parkinson%E2%80%99s-disease-pasadena
  27. #VisualAbstract: Prasinezumab therapy does not affect Parkinson's Disease progression | 2 Minute Medicine, accessed May 8, 2025, https://www.2minutemedicine.com/visualabstract-prasinezumab-therapy-does-not-affect-parkinsons-disease-progression/
  28. Monoclonal antibody Prasinezumab shows promise in slowing rapid Parkinson's progression - News-Medical.net, accessed May 8, 2025, https://www.news-medical.net/news/20240419/Monoclonal-antibody-Prasinezumab-shows-promise-in-slowing-rapid-Parkinsons-progression.aspx
  29. Prasinezumab slows motor progression in Parkinsons disease: beyond the clinical data, accessed May 8, 2025, https://pubmed.ncbi.nlm.nih.gov/39971932/
  30. Parkinson Agent Prasinezumab Misses Primary End Point But Shows Positive Effects in Other Outcomes - NeurologyLive, accessed May 8, 2025, https://www.neurologylive.com/view/parkinson-agent-prasinezumab-misses-primary-end-point-shows-positive-effects-other-outcomes
  31. Intravenous Prasinezumab in Participants With Early Parkinson's Disease - UCSF Clinical Trials, accessed May 8, 2025, https://clinicaltrials.ucsf.edu/trial/NCT04777331
  32. Intravenous Prasinezumab in Early Parkinson's Disease - Cedars-Sinai's clinical trials, accessed May 8, 2025, https://clinicaltrials.cedars-sinai.edu/view/BN42358
  33. AD/PD 2025: Roche highlights time-to-event for assessing prasinezumab in early PD, accessed May 8, 2025, https://www.clinicaltrialsarena.com/analyst-comment/roche-time-to-event-prasinezumab-early-pd/
  34. s201.q4cdn.com, accessed May 8, 2025, https://s201.q4cdn.com/351053094/files/doc_presentations/2025/Jan/10/Prothena-Corporate-Overview-January-10-2025_Final.pdf
  35. Prothena Reports Fourth Quarter and Full Year 2024 Financial Results, and Provides Financial Guidance and Business Highlights - FirstWord Pharma, accessed May 8, 2025, https://firstwordpharma.com/story/5936922
  36. PROTHENA CORP PUBLIC LTD CO SEC 10-K Report - TradingView, accessed May 8, 2025, https://www.tradingview.com/news/tradingview:f489b89346c61:0-prothena-corp-public-ltd-co-sec-10-k-report/
  37. Prothena Fourth Quarter and Year-End 2024 Financial Results, accessed May 8, 2025, https://s201.q4cdn.com/351053094/files/doc_financials/2024/q4/Prothena-FY24-Earnings-Presentation_2-20-25.pdf
  38. Prasinezumab: A Bayesian Perspective on Its Efficacy - PMC - PubMed Central, accessed May 8, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12006898/
  39. FDA Grants Priority Review for Biologics License Application (BLA) and EMA Accepts Marketing Authorisation Application (MAA) for Apitegromab as a Treatment for Spinal Muscular Atrophy - FirstWord Pharma, accessed May 8, 2025, https://firstwordpharma.com/story/5944570

Published at: May 8, 2025

This report is continuously updated as new research emerges.

© Copyright 2025. All Rights Reserved by MedPath