Nerinetide analog NoNO-42 is an investigational synthetic peptide currently under development by NoNO Inc. as a neuroprotective agent primarily targeting acute ischemic stroke (AIS).[1] This compound represents a second-generation therapeutic within NoNO Inc.'s Postsynaptic Density Protein-95 (PSD-95) inhibitor platform, building upon the research and clinical experience gained from its predecessor, nerinetide.[2] The principal driver for the development of NoNO-42 was the identification of a significant limitation with nerinetide: its efficacy was compromised in patients who had received thrombolytic therapy, specifically alteplase.[2] Consequently, NoNO-42 has been engineered for compatibility with thrombolytic agents, a critical advancement aimed at broadening the applicability of PSD-95 inhibition to a larger cohort of AIS patients.[1]
Acute ischemic stroke remains a leading global cause of death and long-term disability, imposing a substantial burden on individuals, families, and healthcare systems.[6] Despite advances in reperfusion therapies such as intravenous thrombolysis and endovascular thrombectomy (EVT), a significant unmet medical need persists for adjunctive neuroprotective treatments that can mitigate ischemic brain injury during the critical early hours following stroke onset. NoNO-42 seeks to fill this therapeutic gap.
The development of NoNO-42 by NoNO Inc., an Ontario-based biotechnology company, reflects a strategic and iterative approach to drug discovery. NoNO Inc. specializes in neurotherapeutics, with a core focus on the PSD-95 protein as a therapeutic target for stroke and other central nervous system (CNS) disorders.[2] The company's strategy centers on selectively inhibiting pathological protein-protein interactions involving PSD-95 that contribute to neuronal damage, while ideally preserving the protein's normal physiological roles.[2] This targeted approach is evident in their pipeline, which includes:
This diversified pipeline underscores NoNO Inc.'s commitment to leveraging the PSD-95 inhibition mechanism across various CNS indications and disease stages. The progression from nerinetide to NoNO-42, in particular, highlights a key aspect of modern pharmaceutical development: the strategic iteration and refinement of drug candidates based on clinical learnings and identified limitations. The challenges encountered with nerinetide, specifically its enzymatic degradation by plasmin when co-administered with alteplase [2], directly catalyzed the engineering of NoNO-42 with enhanced stability. This iterative process, moving from clinical observation of a drug-drug interaction (DDI) back to preclinical peptide engineering and then forward to a new clinical candidate, is a testament to a problem-driven approach to innovation.
NoNO-42, a synthetic peptide, exerts its neuroprotective effects primarily through the inhibition of Postsynaptic Density Protein-95 (PSD-95).[2] PSD-95 is a major scaffolding protein located within the postsynaptic density of excitatory synapses, playing a crucial role in organizing signaling complexes.[14] The therapeutic strategy of NoNO-42 is to selectively disrupt detrimental protein-protein interactions mediated by PSD-95, particularly its coupling with N-methyl-D-aspartate receptors (NMDARs) and neuronal nitric oxide synthase (nNOS), which become pathological during ischemic events.[12] A key design principle is to achieve this inhibition without interfering with the normal physiological functions of these essential neuronal proteins.[2]
A primary mechanism of neuronal injury in AIS is excitotoxicity, triggered by excessive release of the neurotransmitter glutamate. This leads to overactivation of NMDARs, resulting in a massive influx of calcium ions (Ca2+) into neurons. This Ca2+ overload activates a cascade of downstream neurotoxic pathways, including the activation of nNOS, which is physically linked to NMDARs by PSD-95.[12] The subsequent overproduction of nitric oxide (NO) and other reactive oxygen and nitrogen species contributes significantly to neuronal damage and death.[16]
NoNO-42, by inhibiting PSD-95, aims to uncouple NMDARs from nNOS. This uncoupling is hypothesized to prevent the excessive NMDAR activation during ischemia from translating into nNOS-mediated NO overproduction and the ensuing excitotoxic cell death.[12] While some general descriptions might term NoNO-42 a "selective NMDA receptor antagonist" [30], its action is more accurately defined as an indirect modulation of NMDAR-dependent pathological signaling through the specific inhibition of PSD-95's scaffolding role in this pathway. This targeted approach is a significant refinement over traditional, broad-spectrum NMDAR antagonists, which often failed in clinical trials or exhibited prohibitive side effects due to their interference with the essential physiological functions of NMDARs in learning, memory, and normal synaptic transmission.[30] By selectively disrupting a specific pathological linkage (NMDAR-PSD-95-nNOS) that is exacerbated during ischemia, NoNO-42 seeks to offer neuroprotection with an improved therapeutic window.
Neuroinflammation is a well-recognized secondary injury mechanism that significantly contributes to the evolution of brain damage following AIS.[30] Some reports suggest that NoNO-42 may also possess anti-inflammatory properties.[30] These effects could arise indirectly, as a consequence of reduced excitotoxicity and subsequent release of damage-associated molecular patterns (DAMPs) that trigger inflammatory responses. Alternatively, PSD-95 inhibition might have more direct modulatory effects on inflammatory signaling pathways within the brain. However, the primary and most consistently emphasized mechanism of action for PSD-95 inhibitors like NoNO-42 is the attenuation of excitotoxicity. The extent and directness of any anti-inflammatory effects require further specific investigation.
As an analog of nerinetide, NoNO-42 shares the same fundamental pharmacological target (PSD-95) and the core mechanism of inhibiting its pathological interactions to reduce excitotoxicity.[1] The critical pharmacological distinction between nerinetide and NoNO-42 lies not in their primary target engagement but in NoNO-42's engineered resistance to enzymatic degradation by plasmin. This modification is designed to ensure its stability and efficacy when co-administered with thrombolytic agents such as alteplase, which activate plasmin.[2]
The development of NoNO-42 underscores the importance of peptide engineering in creating viable CNS therapeutics. Peptides offer high target specificity but often face challenges with in vivo stability and penetration of the blood-brain barrier (BBB). Nerinetide (Tat-NR2B9c) incorporated the HIV-Tat protein transduction domain to enhance cell penetration.[11] NoNO-42 further refines this approach by incorporating modifications, likely the substitution of L-amino acids with D-amino acids at plasmin-sensitive cleavage sites, to confer resistance to enzymatic degradation in the bloodstream.[4] This advanced peptide chemistry is crucial for overcoming the pharmacokinetic limitations that hindered nerinetide's utility in a significant portion of the stroke patient population.
The potential for PSD-95 inhibitors to offer not only acute neuroprotection but also contribute to longer-term neurorestoration is an intriguing prospect. PSD-95 is a cornerstone of synaptic structure and plasticity.[14] Modulating its function could, beyond limiting acute damage, influence the brain's capacity for reorganization and functional recovery in the chronic phase post-stroke. This broader therapeutic horizon is supported by NoNO Inc.'s development of NoNO-SC, a subcutaneous formulation of a PSD-95 inhibitor, for indications such as stroke recovery and dementia.[2]
The development of NoNO-42 is directly rooted in the clinical experience and identified limitations of NoNO Inc.'s first-generation PSD-95 inhibitor, nerinetide. Understanding nerinetide's journey is crucial to appreciating the strategic rationale behind NoNO-42.
Nerinetide (NA-1, Tat-NR2B9c) showed initial promise in preclinical models and early clinical studies.[10] Its efficacy was further investigated in several large-scale Phase 3 trials:
The consistent theme emerging from these trials was that nerinetide's potential benefits were masked or negated in the presence of alteplase and were most apparent with early administration in patients undergoing reperfusion.
The most significant challenge identified for nerinetide was its interaction with alteplase. Pharmacokinetic analyses from the ESCAPE-NA1 trial revealed that co-administration of alteplase led to an approximate 60% reduction in nerinetide plasma concentrations.[21] This occurs because alteplase, a thrombolytic drug, activates plasmin. Plasmin, a serine protease, was found to proteolytically cleave the nerinetide peptide, thereby inactivating it and diminishing its neuroprotective efficacy.[4] This DDI effectively limited nerinetide's utility to patients not receiving alteplase, a substantial portion of the AIS population targeted for acute intervention.
NoNO-42 was specifically developed as a "next-generation" PSD-95 inhibitor engineered to be resistant to plasmin-mediated degradation.[2] This crucial modification is intended to allow NoNO-42 to maintain its structural integrity and pharmacological activity even when co-administered with thrombolytic agents like alteplase.[1] The primary goal of this design is to extend the potential neuroprotective benefits of PSD-95 inhibition to all eligible AIS patients, including the approximately 10% who receive thrombolytics and in whom nerinetide's efficacy was compromised.[1]
The development of NoNO-42, therefore, is not merely an incremental improvement but a targeted solution to a well-defined clinical problem identified through rigorous Phase 3 testing of its predecessor. This iterative approach, where clinical challenges inform preclinical drug design to yield improved next-generation therapeutics, is a hallmark of rational drug development. The ability of NoNO-42 to function alongside standard-of-care thrombolytics would significantly simplify treatment algorithms in the hyperacute stroke setting and potentially make neuroprotection a viable option for a much wider patient base. The positive signals observed in specific nerinetide subgroups (early treatment, reperfusion therapy recipients) provide a strong rationale for NoNO-42, assuming its plasmin resistance translates to maintained efficacy in alteplase-treated patients.
The following table provides a comparative overview of nerinetide and NoNO-42:
Table 1: Comparison of Nerinetide and Nerinetide Analog NoNO-42
Feature | Nerinetide | Nerinetide Analog NoNO-42 |
---|---|---|
Generation | First-generation PSD-95 inhibitor | Next-generation PSD-95 inhibitor |
Primary Pharmacological Target | Postsynaptic Density Protein-95 (PSD-95) | Postsynaptic Density Protein-95 (PSD-95) |
Key Mechanism of Action | Inhibition of PSD-95 interaction with NMDAR-nNOS, reducing excitotoxicity | Inhibition of PSD-95 interaction with NMDAR-nNOS, reducing excitotoxicity |
Susceptibility to Plasmin/Alteplase | Yes, cleaved by plasmin leading to reduced efficacy when co-administered with alteplase 4 | Designed for plasmin resistance, intended for use with or without thrombolytics 1 |
Primary Indication Focus | Acute Ischemic Stroke in patients undergoing EVT not receiving alteplase 2 | Acute Ischemic Stroke in patients eligible for thrombolysis +/- EVT 1 |
Key Clinical Trials | ESCAPE-NA1 (NCT02930018), ESCAPE-NEXT (NCT04462536), FRONTIER (NCT02315443) 9 | NCT05636306 (Phase 1), ACT-42 (NCT06403267, Phase 2b), ACT-GLOBAL (NCT06352632, Phase 3 platform) 2 |
Current Highest Development Stage | Phase 3 (completed/ongoing for specific populations) 10 | Phase 2b/3 (ongoing) 1 |
Key Differentiator/Development Goal | Demonstrate neuroprotection in alteplase-naive EVT patients | Demonstrate neuroprotection in broader AIS population, including alteplase-treated patients; enable earlier admin. |
Formulation Goal | Intravenous | Intravenous, potential for StrokePen autoinjector 2 |
This comparative framework underscores the targeted evolution of NoNO-42, aiming to build upon the mechanistic understanding gained from nerinetide while addressing its primary clinical limitation.
The preclinical development of NoNO-42 was heavily influenced by the need to create a PSD-95 inhibitor that retains neuroprotective efficacy in the presence of thrombolytic agents like alteplase.
The foundational principle of PSD-95 inhibition as a neuroprotective strategy has been established through numerous preclinical studies, primarily with nerinetide. These studies, conducted in various animal models of acute ischemic stroke, consistently demonstrated that inhibiting PSD-95 within a critical time window (typically within 3 hours of stroke onset) can reduce infarct volume and improve neurological outcomes.[8] NoNO-42, being an analog of nerinetide, inherits this preclinical rationale.[1] Early preclinical investigations specific to NoNO-42 also indicated promising neuroprotective activity.[30]
The pivotal challenge that NoNO-42's preclinical program sought to overcome was nerinetide's susceptibility to proteolytic degradation by plasmin, an enzyme whose activity is significantly increased upon administration of alteplase.[4] Research efforts focused on modifying the peptide structure of nerinetide to render it resistant to this enzymatic cleavage without compromising its ability to inhibit PSD-95.
A seminal study published in Science Translational Medicine, titled "Plasmin-resistant PSD-95 inhibitors resolve effect-modifying drug-drug interactions between alteplase and nerinetide in acute stroke," detailed the successful development of such protease-resistant compounds.[4] This research, originating from the same group involved in nerinetide's development, including Dr. Michael Tymianski, showed that nerinetide could maintain its effectiveness if administered before alteplase in a rat model of embolic middle cerebral artery occlusion (eMCAO). This was attributed to nerinetide's short plasma half-life, allowing it to exert its effect before substantial plasmin activation by subsequently administered alteplase.[14]
More critically for the development of NoNO-42, the Science Translational Medicine study demonstrated that specific amino acid modifications could confer plasmin resistance. By substituting certain L-amino acids (the natural enantiomeric form) at or near plasmin cleavage sites with their D-enantiomeric counterparts, the peptide's stability in the presence of plasmin was significantly enhanced. A lead compound from this work, termed d-Tat-l-2B9c (where 'd-Tat' signifies the Tat peptide with D-amino acids and 'l-2B9c' the NR2B9c inhibitory peptide largely in L-form), was shown to eliminate protease sensitivity and maintain neuroprotective effectiveness in the rat eMCAO model, even when alteplase was present.[14]
Further insights into these modifications are provided in patent EP4106818A1, which lists Dr. Michael Tymianski as an inventor.[12] The patent describes methods for enhancing plasmin resistance in PSD-95 inhibitor peptides by incorporating D-amino acids. The strategy often involves placing D-amino acids immediately C-terminal to basic residues (arginine or lysine), which are preferred cleavage sites for plasmin. The patent also mentions other strategies, such as the use of retro-inverso peptides (e.g., RI-NA-1), where the amino acid sequence is reversed, and all amino acids are in the D-configuration to mimic the side-chain topology of the original L-peptide.[12]
NoNO-42 is the clinical candidate that has emerged from this dedicated research into plasmin-resistant PSD-95 inhibitors.[2] Its design for co-administration with thrombolytic agents directly reflects the successful preclinical proof-of-concept established by compounds like d-Tat-l-2B9c.[1] While the exact sequence of NoNO-42 may not be publicly disclosed as identical to d-Tat-l-2B9c, the scientific rationale and the timing of its development strongly suggest it incorporates these principles of D-amino acid substitution or similar peptide engineering strategies to achieve plasmin resistance.
This translational research pathway—from identifying a clinical DDI with nerinetide, to targeted preclinical research yielding plasmin-resistant analogs, and culminating in the clinical development of NoNO-42—is a clear example of problem-solving in drug development. The innovation in NoNO-42 lies significantly in this sophisticated peptide engineering, which enhances its pharmacokinetic profile (resistance to enzymatic degradation) and thereby expands its potential clinical utility to a broader stroke patient population.
While NoNO Inc. has stated that success with nerinetide trials (in alteplase-naive patients) would substantially de-risk NoNO-42 development due to their shared PSD-95 inhibitory mechanism [2], it is important to recognize that NoNO-42 is a new molecular entity. The modifications conferring plasmin resistance, while intended to improve its profile, necessitate independent clinical validation of its overall safety, pharmacokinetics, and efficacy, particularly in the alteplase-positive population it is designed to serve. The de-risking primarily applies to the biological plausibility of PSD-95 inhibition as a neuroprotective strategy, which nerinetide's data has supported under specific conditions.
The clinical development of NoNO-42 is progressing through a structured program, starting with Phase 1 safety and pharmacokinetic studies in healthy volunteers, and moving towards efficacy assessments in acute ischemic stroke patients in later-phase trials.
NoNO-42 is currently being evaluated in later-stage clinical trials, notably as part of a larger adaptive platform trial.
The clinical development strategy for NoNO-42 reflects several contemporary trends in trial design. The use of adaptive platform trials like ACT-GLOBAL allows for more efficient evaluation of multiple therapeutic candidates and combinations, which is particularly advantageous in complex and heterogeneous conditions such as stroke.[2] This approach can accelerate the identification of effective treatments and responsive patient populations. Furthermore, the patient selection criteria for ACT-42, focusing on early intervention (within 3 hours) and patients eligible for thrombolysis, directly incorporates the key learnings from the extensive nerinetide clinical program, aiming to test NoNO-42 in a population where PSD-95 inhibition is hypothesized to confer the most significant benefit, now with the added advantage of alteplase compatibility. The potential discrepancy in blinding (PROBE vs. double-blind) for ACT-42 might reflect different reporting perspectives or specific phases/sub-studies within the adaptive design; however, a blinded assessment of endpoints remains a cornerstone for robust efficacy evaluation.
Table 2: Summary of NoNO-42 Clinical Trials
NCT ID | Phase | Official Title | Status | Target Population | Intervention (Drug & Dosage) | Key Primary Endpoint(s) | Key Secondary Endpoints | Sponsor(s)/Key Collaborator(s) |
---|---|---|---|---|---|---|---|---|
NCT05636306 2 | Phase 1A | A Phase 1, Randomized, Placebo Controlled, Double-Blind, Single Dose Escalation Study in Healthy Adults | Completed | Healthy Adult Volunteers | Single ascending IV doses of NoNO-42 vs. Placebo | Safety and Tolerability (e.g., adverse events) | Pharmacokinetics of NoNO-42 | NoNO, Inc. |
NCT06403267 (ACT-42) 22 | Phase 2b | A Multicentre, Randomized, Double-blinded, Placebo-controlled, Parallel Group, Single-dose Design to Determine the Efficacy and Safety of NoNO-42 in Participants with AIS... | Active, Recruiting | AIS patients (45-90 yrs) eligible for thrombolysis +/- EVT, within 3 hrs of onset | Single 2.6 mg/kg IV dose of NoNO-42 or Placebo (20-min inf) | Reduction in global disability (shift on mRS at Day 90) | Excellent functional outcome (mRS 0-1 at Day 90); Reduced stroke worsening; 90-day mortality; Health-related QoL (EQ-5D-5L at Day 90) | NoNO, Inc., University of Calgary, McGill University Health Centre |
NCT06352632 (ACT-GLOBAL) 2 | Phase 3 (Platform) | Adaptive Platform Trial for Stroke to Identify Candidate Therapies for a Favorable Outcome | Active, Recruiting | Acute Ischemic Stroke Patients | Multiple arms, including NoNO-42 via ACT-42 domain | Improvement in functional outcome (e.g., mRS) | Various, depending on specific interventions being tested within the platform | The George Institute for Global Health, University of Calgary |
AIS: Acute Ischemic Stroke; EVT: Endovascular Thrombectomy; IV: Intravenous; mRS: modified Rankin Scale; QoL: Quality of Life.
The primary therapeutic focus for Nerinetide analog NoNO-42 is acute ischemic stroke (AIS), a condition with a pressing need for effective neuroprotective interventions.
NoNO-42 is being developed for the treatment of all individuals experiencing an AIS, with a particular emphasis on its utility in conjunction with standard reperfusion therapies.[2] Clinical trials, such as ACT-42, are specifically enrolling AIS patients who are eligible for intravenous thrombolysis (with agents like alteplase) and/or endovascular thrombectomy, and who can be treated within a narrow therapeutic window, typically 3 hours from symptom onset or last known well time.[22]
A cornerstone of NoNO-42's therapeutic potential is its engineered resistance to plasmin, making it compatible with thrombolytic drugs.[1] This is a significant advancement over its predecessor, nerinetide, whose efficacy was diminished in patients receiving alteplase. By overcoming this limitation, NoNO-42 aims to extend the benefits of PSD-95 inhibition to a broader segment of the AIS population, including the approximately 10% of patients who receive thrombolytics and might not have benefited from nerinetide.[1] This compatibility simplifies treatment decisions in the hyperacute stroke setting, as NoNO-42 could potentially be administered without needing to consider a patient's thrombolytic status.
While the immediate clinical development of NoNO-42 is centered on AIS, the underlying mechanism of PSD-95 inhibition has implications for a wider range of neurological and neurodegenerative conditions where excitotoxicity and synaptic dysfunction play a role. Some sources suggest that NoNO-42, by targeting NMDA receptor-mediated pathways and potentially exerting anti-inflammatory effects, could have therapeutic potential in conditions such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis (ALS).[30]
NoNO Inc.'s broader pipeline supports this vision. The company explicitly mentions the potential of their PSD-95 inhibitor platform for traumatic brain injury and neuropathic pain, in addition to stroke and Alzheimer's disease.[4] Furthermore, the development of NoNO-SC, a subcutaneously injectable PSD-95 inhibitor, is targeted towards subacute and chronic neurological conditions, including stroke recovery, vascular dementia, and Alzheimer's disease.[2] This suggests a long-term strategy to leverage PSD-95 modulation across a spectrum of CNS disorders. However, it is important to note that active clinical trials for NoNO-42 are currently focused on AIS; its application in other neurodegenerative diseases remains largely in the exploratory or preclinical stages for the platform technology.
A particularly innovative aspect of NoNO-42's development is its association with the "StrokePen" program.[2] This initiative aims to formulate NoNO-42 for administration via an autoinjector. Such a device could enable rapid, out-of-hospital administration by caregivers, paramedics, or upon arrival at an emergency department, even before definitive neuroimaging or specialized stroke unit care.[2]
The rationale for the StrokePen is compelling: the efficacy of neuroprotective agents is critically time-dependent, with the greatest benefit observed when administered as early as possible after stroke onset.[7] By facilitating pre-hospital or immediate emergency room administration, the StrokePen could significantly shorten the time-to-treatment, potentially "buying time" for brain cells and improving outcomes before or during transport to a specialized stroke center and initiation of reperfusion therapies. This approach could represent a paradigm shift in acute stroke management, moving the first line of neuroprotective defense closer to the patient at the point of initial contact.
However, the deployment of such a pre-hospital intervention also presents challenges. Accurate stroke diagnosis outside of a hospital setting is difficult, as highlighted by the FRONTIER trial with nerinetide, which included a significant number of "stroke mimics".[7] Therefore, the safety profile of NoNO-42 in individuals who are not actually experiencing an ischemic stroke would be a critical determinant for the feasibility and regulatory acceptance of widespread pre-hospital use via the StrokePen.
As an investigational compound, NoNO-42 is navigating the rigorous regulatory pathways required for new drug approval.
The provided research materials do not indicate that NoNO-42 has received specific regulatory designations such as Orphan Drug Designation or Fast Track status from the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA) at this time.[43] While its predecessor, nerinetide, did receive Orphan Drug Designation from the FDA for subarachnoid hemorrhage and acute ischemic stroke (FDA Designations 377812 and 412713) [11], these designations are specific to the molecular entity and indication and do not automatically transfer to an analog like NoNO-42. Regulatory agencies will evaluate NoNO-42 based on its own merits and clinical trial data. Currently, NoNO-42 is advancing through late-stage clinical development, with the ACT-42 trial operating as a Phase 2b component within the larger Phase 3 ACT-GLOBAL adaptive platform trial.[1]
Key future milestones for the NoNO-42 program include:
The successful development of NoNO-42 could have a profound impact on the management of acute ischemic stroke. If proven safe and effective, it could become the first neuroprotective agent broadly compatible with standard-of-care reperfusion therapies, including thrombolytics. This would address a significant limitation of previous neuroprotective candidates and simplify treatment algorithms in the hyperacute setting.
Furthermore, the StrokePen concept, enabling very early, potentially pre-hospital administration, holds the promise of revolutionizing the initial approach to stroke care.[2] By extending the window for effective neuroprotection closer to the onset of symptoms, NoNO-42 could significantly improve long-term functional outcomes for stroke survivors.
However, the path to approval for neuroprotective agents in stroke is notoriously challenging. The heterogeneity of stroke pathology, the narrow therapeutic window, and the complexities of conducting trials in an emergency setting contribute to a high attrition rate for investigational drugs in this field. The overall "neutral" results from the large nerinetide trials, despite promising signals in specific subgroups, underscore this difficulty.[7] NoNO-42 will undoubtedly face rigorous scrutiny from regulatory authorities, who will require robust and statistically significant evidence of clinical benefit from well-designed and adequately powered Phase 3 trials.
The use of adaptive trial designs, such as in ACT-GLOBAL and ACT-42, may help in prospectively identifying patient subgroups most likely to respond to NoNO-42, potentially through biomarker or advanced imaging stratification.[2] This data-driven approach, building on the lessons learned from nerinetide, could be crucial for demonstrating a clear and clinically meaningful benefit. The strong international collaboration evident in the NoNO-42 clinical program is also a positive factor, facilitating patient recruitment and enhancing the generalizability of trial results, which are vital for global regulatory acceptance.[2]
Nerinetide analog NoNO-42 represents a focused and scientifically driven effort to advance neuroprotection for acute ischemic stroke. Developed by NoNO Inc., this synthetic peptide builds upon the mechanistic understanding of PSD-95 inhibition gained from its predecessor, nerinetide, while specifically addressing a key limitation: incompatibility with thrombolytic therapy due to plasmin-mediated degradation. Through innovative peptide engineering aimed at conferring plasmin resistance, NoNO-42 is designed to be effective in a broader population of AIS patients, including those receiving alteplase.
The ongoing clinical development program, notably the Phase 2b ACT-42 trial within the Phase 3 ACT-GLOBAL adaptive platform, will be critical in determining the safety and efficacy of NoNO-42. The trial design incorporates lessons from the nerinetide program, focusing on early administration (within 3 hours of onset) in patients selected for reperfusion therapies. Successful outcomes from these trials could establish NoNO-42 as a valuable adjunctive therapy in the hyperacute management of stroke.
The potential development of the StrokePen autoinjector formulation for NoNO-42 further underscores the ambition to shift the paradigm of stroke care towards earlier intervention, potentially even in the pre-hospital setting. This could significantly expand the therapeutic window for neuroprotection.
Despite the promising rationale and innovative design, the path for neuroprotective agents in stroke remains challenging. The heterogeneity of the condition and the complexities of acute care trials necessitate robust and unequivocal evidence of clinical benefit. NoNO-42's journey reflects a pragmatic approach to innovation, leveraging past clinical experiences to refine a therapeutic strategy. If successful, NoNO-42 could offer a significant advancement in addressing the vast unmet medical need for effective treatments that reduce disability and improve outcomes for stroke patients worldwide. The medical and scientific communities await the results of the ongoing late-stage trials with considerable interest.
References
1 Ischemic Stroke - Pipeline Insight, 2025. DelveInsight.
9 Efficacy and safety of intravenous nerinetide initiated by paramedics in the field for acute cerebral ischaemia within 3 h of symptom onset (FRONTIER): a phase 2, multicentre, randomised, double-blind, placebo-controlled study. PubMed, NCBI.
30 What is NoNO-42 used for? Patsnap Synapse.
2 NoNO-42 Drug Profile. Patsnap Synapse.
2 NoNO-42 Drug Profile (duplicate). Patsnap Synapse.
7 UCalgary stroke researchers find silver linings in neuroprotection studies. Cumming School of Medicine, University of Calgary.
10 Nerinetide - Arbor Vita Corporation/NoNO. AdisInsight, Springer.
11 Nerinetide Drug Information. NCATS Inxight Drugs.
30 What is NoNO-42 used for? (duplicate). Patsnap Synapse.
47 NONO couples the circadian clock to the cell cycle. PNAS.
22 A Multicentre, Randomized, Double-blinded, Placebo-controlled, Parallel Group, Single-dose Design to Determine the Efficacy and Safety of NoNO-42 in Participants with Acute Ischemic Stroke Selected for Thrombolysis. CIHR Funding Decisions Database.
23 ACT-GLOBAL Adaptive Platform Trial for Stroke. Larvol Delta.
24 NoNO, Inc. Company Profile. Patsnap Synapse.
4 NoNO publishes next-gen PSD-95 inhibitors extending the therapeutic potential of PSD-95 inhibition in acute stroke. NoNO Inc. Press Release. (2021-04-07).
4 NoNO publishes next-gen PSD-95 inhibitors extending the therapeutic potential of PSD-95 inhibition in acute stroke (duplicate). NoNO Inc. Press Release. (2021-04-07).
12 Patent EP4106818A1: Inhibition of reperfusion injury with a PSD-95 inhibitor. Google Patents.
30 What is NoNO-42 used for? (duplicate). Patsnap Synapse.
7 UCalgary stroke researchers find silver linings in neuroprotection studies (duplicate). Cumming School of Medicine, University of Calgary.
15 Predictors of Clinical Outcome in Patients With Acute Ischemic Stroke Not Treated With Alteplase in the ESCAPE-NA1 Trial. Journal of Stroke.
25 CIHR Clinical Trials Projects – Second Round of Funding. Government of Canada.
13 Disappointing Results for Nerinetide in Stroke. Medscape.
26 NoNO announces first patient dosed with NoNO-42 in Phase 1 study. Neuronews International.
48 NONO Is a Negative Regulator of SOX2 Promoter. PMC, NCBI. (Note: This snippet appears unrelated to NoNO-42 the drug).
27 NoNO-42 Trial in Acute Ischemic Stroke Patients Selected for Thrombolysis with or Without Endovascular Thrombectomy. CenterWatch.
12 Patent EP4106818A1: Inhibition of reperfusion injury with a PSD-95 inhibitor (duplicate). Google Patents.
14 Plasmin-resistant PSD-95 inhibitors resolve effect-modifying drug-drug interactions between alteplase and nerinetide in acute stroke. Science Translational Medicine, PubMed.
8 Protecting the Brain Against Stroke. UHN Research.
7 UCalgary stroke researchers find silver linings in neuroprotection studies (duplicate). Cumming School of Medicine, University of Calgary.
15 Predictors of Clinical Outcome in Patients With Acute Ischemic Stroke Not Treated With Alteplase in the ESCAPE-NA1 Trial (duplicate). Journal of Stroke.
27 NoNO-42 Trial in Acute Ischemic Stroke Patients Selected for Thrombolysis with or Without Endovascular Thrombectomy (duplicate). CenterWatch.
15 Predictors of Clinical Outcome in Patients With Acute Ischemic Stroke Not Treated With Alteplase in the ESCAPE-NA1 Trial (duplicate). Journal of Stroke.
27 NoNO-42 Trial in Acute Ischemic Stroke Patients Selected for Thrombolysis with or Without Endovascular Thrombectomy (duplicate). CenterWatch.
5 NoNO Inc. Announces First Patient Dosed with NoNO-42 in a First in Humans Phase 1 Study. NoNO Inc. Press Release. (2022-11-22).
16 Toronto’s NoNO Reports Major Results in Stroke Phase III Trial. BioSpace.
17 Nerinetide Reduces Infarct Growth in Patients With Ischemic Stroke Not Treated With Alteplase. Stroke: Vascular and Interventional Neurology, AHA Journals.
18 Plasmin-resistant PSD-95 inhibitors resolve effect-modifying drug-drug interactions between alteplase and nerinetide in acute stroke. ResearchGate. (Abstract of Sci Transl Med paper).
7 UCalgary stroke researchers find silver linings in neuroprotection studies (duplicate). Cumming School of Medicine, University of Calgary.
49 Problems in the exemplar trials include failing to define the subgroup in advance or even changing its definition... PMC, NCBI. (General discussion on clinical trial subgroups).
43 FDA Administrative Correspondence for NDA 210922 (Patisiran). FDA. (Unrelated to NoNO-42).
44 FDA Post-Approval Studies Database Information. FDA. (General FDA database info, unrelated).
3 Ischemic Stroke- Pipeline Insight, 2025 (duplicate). Research and Markets.
6 NoNO Inc. Pipeline Information. NoNO Inc. Website.
40 Clinical Trials Information System designated as WHO primary registry. EMA News.
41 WHO International Clinical Trials Registry Platform (ICTRP). WHO.
42 Global analysis of the WHO International Clinical Trials Registry Platform (ICTRP) database. PMC, NCBI.
38 Difference Between Primary & Secondary Clinical Trial Endpoints. Atlantia Clinical Trials Blog.
39 What are primary vs. secondary outcomes? Evidence For Action FAQ.
16 Toronto’s NoNO Reports Major Results in Stroke Phase III Trial (duplicate). BioSpace.
31 Long-term safety and efficacy of efgartigimod in generalized myasthenia gravis: results from the ADAPT+ open-label extension study. PMC, NCBI. (Unrelated to NoNO-42).
28 A Multicentre, Prospective, Randomized, Open Label, Blinded-Endpoint, Placebo-controlled, Single-dose Trial to Determine the Efficacy and Safety of NoNO-42... (ACT-42 Trial). ClinicalTrial.be.
27 NoNO-42 Trial in Acute Ischemic Stroke Patients Selected for Thrombolysis with or Without Endovascular Thrombectomy (duplicate). CenterWatch.
29 NoNO 42 Drug Profile. AdisInsight, Springer.
19 Nerinetide Reduces Infarct Growth in Patients With Ischemic Stroke Not Treated With Alteplase (duplicate). Stroke: Vascular and Interventional Neurology, AHA Journals.
50 Hydroxychloroquine Alleviates Ischemic Stroke Pathology by Inhibiting Microglial Pyroptosis. eNeuro. (Unrelated to NoNO-42).
45 Fast Track Status Given to Givinostat for Polycythemia Vera. OncNursingNews. (Unrelated to NoNO-42).
46 Oncology Drugs Fast-Tracked by the FDA in February 2025. Oncology News Central. (Unrelated to NoNO-42).
32 Patient-reported outcome endpoints in clinical trials of novel type 2 diabetes treatments: a systematic literature review. PubMed, NCBI. (General clinical trial endpoint info).
33 ClinicalTrials.gov Glossary. ClinicalTrials.gov. (General clinical trial definitions).
28 A Multicentre, Prospective, Randomized, Open Label, Blinded-Endpoint, Placebo-controlled, Single-dose Trial to Determine the Efficacy and Safety of NoNO-42... (ACT-42 Trial) (duplicate). ClinicalTrial.be.
20 Nerinetide Reduces Infarct Growth in Patients With Ischemic Stroke Not Treated With Alteplase (duplicate). Stroke: Vascular and Interventional Neurology, AHA Journals.
34 Endovascular thrombectomy alone versus intravenous thrombolysis plus endovascular thrombectomy for acute ischaemic stroke: a systematic review and meta-analysis. Journal of NeuroInterventional Surgery, BMJ. (General stroke trial methodology).
35 ISC 2018 Ongoing Clinical Trial Abstracts. AHA Journals. (General stroke trial abstracts).
36 AANEM Abstracts Guide. AANEM. (General abstract guidelines).
37 Abstracts from the 20th IPEG Annual Congress for Endosurgery in Children. PMC, NCBI. (Unrelated abstracts).
25 CIHR Clinical Trials Projects – Second Round of Funding (duplicate). Government of Canada.
6 NoNO Inc. Pipeline Information (duplicate). NoNO Inc. Website.
2 NoNO-42 Drug Profile (duplicate). Patsnap Synapse.
29 NoNO 42 Drug Profile (duplicate). AdisInsight, Springer.
28 A Multicentre, Prospective, Randomized, Open Label, Blinded-Endpoint, Placebo-controlled, Single-dose Trial to Determine the Efficacy and Safety of NoNO-42... (ACT-42 Trial) (duplicate). ClinicalTrial.be.
5 NoNO Inc. Announces First Patient Dosed with NoNO-42 in a First in Humans Phase 1 Study (duplicate). NoNO Inc. Press Release. (2022-11-22).
4 NoNO publishes next-gen PSD-95 inhibitors... (duplicate). NoNO Inc. Press Release. (2021-04-07).
21 Reports Groundbreaking Results from the Phase 3 ESCAPE-NA1 Study... NoNO Inc. Press Release. (2020-02-20).
4 NoNO publishes next-gen PSD-95 inhibitors....4 NoNO Inc. Press Release.
2 NoNO-42 Drug Profile.2 Patsnap Synapse.
1 Ischemic Stroke - Pipeline Insight, 2025.1 DelveInsight.
22 CIHR Funding Decisions Database (general page). CIHR.
7 UCalgary stroke researchers find silver linings....7 Cumming School of Medicine, University of Calgary.
12 Patent EP4106818A1.12 Google Patents.
14 Plasmin-resistant PSD-95 inhibitors....14 PubMed, NCBI.
6 NoNO Inc. Pipeline Information.6 NoNO Inc. Website.
Published at: May 14, 2025
This report is continuously updated as new research emerges.