MedPath

Mitiperstat Advanced Drug Monograph

Published:Oct 24, 2025

Generic Name

Mitiperstat

Report on the Investigational Myeloperoxidase Inhibitor: Mitiperstat (AZD4831)

Section 1: Executive Summary

Mitiperstat, also known as AZD4831, is a first-in-class, orally available, irreversible inhibitor of the enzyme myeloperoxidase (MPO) developed by AstraZeneca.[1] The therapeutic rationale for its development was predicated on a compelling biological hypothesis: that by inhibiting MPO, a key enzyme released by neutrophils during inflammation, it could mitigate the downstream effects of oxidative stress and microvascular dysfunction believed to drive the pathophysiology of complex inflammatory conditions.[3] The primary focus of its clinical program was heart failure with preserved or mildly reduced ejection fraction (HFpEF/HFmrEF), with parallel investigations in chronic obstructive pulmonary disease (COPD) and non-alcoholic steatohepatitis (NASH).[3]

The clinical journey of Mitiperstat followed a path from initial promise to definitive failure. The Phase 2a SATELLITE trial successfully demonstrated proof of mechanism, confirming that Mitiperstat achieved robust target engagement by significantly reducing MPO activity in patients with HFpEF.[7] This success, coupled with a generally acceptable safety profile, supported its advancement into a large-scale, pivotal trial. However, the subsequent seamless Phase 2b/3 ENDEAVOR trial, involving over 700 patients, failed to meet both of its co-primary endpoints. Mitiperstat did not produce any statistically significant improvement in patient-reported symptoms, as measured by the Kansas City Cardiomyopathy Questionnaire (KCCQ), or in functional capacity, as measured by the 6-minute walk distance (6MWD), when compared to placebo.[9]

Throughout its development, Mitiperstat demonstrated a generally tolerable safety profile. The most consistently observed treatment-related adverse event was a self-limited maculopapular rash.[7] Importantly, dedicated cardiac safety studies confirmed that Mitiperstat does not pose a risk of QT interval prolongation, a critical safety attribute for any cardiovascular therapeutic.[11] Despite its tolerability and proven pharmacological activity, the lack of translation into clinical efficacy in the ENDEAVOR trial was a decisive setback. While exploratory analyses suggested a numerical, non-significant trend towards a reduction in heart failure hospitalizations, this signal was insufficient to salvage the program.[9]

Ultimately, in early 2025, AstraZeneca announced the discontinuation of the Mitiperstat programs for heart failure and NASH, citing "strategic portfolio prioritization".[13] This decision reflects the high bar for demonstrating clinical value in a competitive and evolving therapeutic landscape. Mitiperstat's story serves as a significant case study in drug development, illustrating the profound challenge of translating a potent and selective mechanism of action against a plausible biological target into meaningful clinical benefits for patients.

Table 1: Summary of Key Mitiperstat Clinical Trials

Trial Name (Identifier)PhaseIndicationNumber of PatientsPrimary Endpoint(s)Key Outcome
SATELLITE (NCT03756285)2aHFpEF/HFmrEF41MPO specific activity (target engagement) and safetyPositive: Successfully demonstrated >50% MPO inhibition; well tolerated. Efficacy exploratory due to early termination.7
ENDEAVOR (NCT04986202)2b/3HFpEF/HFmrEF711Change in KCCQ-TSS and 6MWD at 16 weeksNegative: Failed to meet both co-primary endpoints; no significant improvement in symptoms or exercise capacity.9
CRESCENDO (NCT05492877)2aModerate-to-severe COPD~406Time to first composite exacerbation (CompEx) eventResults Not Public: Trial completed in August 2024, but final results have not been released.14

Section 2: Molecular Profile and Pharmacological Characteristics

2.1 Chemical Identity and Properties

Mitiperstat (AZD4831) is classified as a new molecular entity and a small molecule drug belonging to several chemical classes, including amines, chlorophenols, pyrimidinones, and pyrroles.[2] It was developed internally by AstraZeneca.[8] As an investigational compound, it is identified by a comprehensive set of chemical and registry identifiers that provide a unique molecular fingerprint.

  • Primary Synonym: AZD4831 [1]
  • IUPAC Name: 1--4-chlorophenyl]methyl]-2-sulfanylidene-5H-pyrrolo[3,2-d]pyrimidin-4-one [1]
  • Chemical Formula: $C_{15}H_{15}ClN_{4}OS$ [1]
  • Molar Mass: 334.82 g·mol−1 [1]
  • CAS Number: 1933460-19-5 [1]
  • PubChem CID: 121362450 [1]
  • ChEMBL ID: CHEMBL5095218 [1]
  • UNII: S6GYK3X4QQ [1]
  • DrugBank ID: DB18963 [18]
  • InChI: InChI=1S/C15H15ClN4OS/c1-8(17)11-6-10(16)3-2-9(11)7-20-12-4-5-18-13(12)14(21)19-15(20)22/h2-6,8,18H,7,17H2,1H3,(H,19,21,22)/t8-/m1/s1 [1]
  • SMILES: CC@HN

2.2 Mechanism of Action and Target Selectivity

Mitiperstat is an orally effective, mechanism-based, irreversible inhibitor of myeloperoxidase. Its mechanism of action is highly specific and durable. Rather than simply binding to the enzyme's active site, Mitiperstat acts as a substrate for MPO. During the enzymatic reaction, Mitiperstat is processed into a reactive intermediate that forms a permanent, covalent bond with the heme prosthetic group within the MPO enzyme. This covalent modification results in the irreversible inactivation of the enzyme's catalytic activity. This "suicide inhibition" mechanism ensures a prolonged pharmacodynamic effect that can outlast the drug's presence in plasma.

The potency and selectivity of Mitiperstat for its primary target are defining features of its pharmacological profile. In vitro assays have quantified its high affinity for MPO and its significantly lower affinity for other related enzymes:

  • Myeloperoxidase (MPO): Half-maximal inhibitory concentration ($IC_{50}$) of 1.5 nM.
  • Thyroid Peroxidase (TPO): $IC_{50}$ of 0.69 μM (690 nM).
  • Cytochrome P450 3A4 (CYP3A4): Weak inhibition with an $IC_{50}$ of 6 μM (6000 nM).

The substantial difference between the concentration required to inhibit MPO and the concentrations needed to affect off-targets like TPO and CYP3A4 is a critical pharmacological attribute. The drug is approximately 460-fold more selective for MPO than for TPO, and 4000-fold more selective for MPO than for CYP3A4. This large potency gap creates a favorable therapeutic window. It strongly suggests that at the low nanomolar concentrations required to achieve therapeutic inhibition of MPO, Mitiperstat is highly unlikely to cause clinically significant off-target effects. Specifically, the low affinity for TPO minimizes the risk of disrupting thyroid hormone synthesis, a common concern for peroxidase inhibitors. Similarly, the weak inhibition of CYP3A4, a major enzyme in drug metabolism, indicates a low potential for clinically relevant drug-drug interactions. From a drug development perspective, this high degree of selectivity is a desirable characteristic that de-risks the compound from predictable, mechanism-based toxicities.

Section 3: Clinical Pharmacology: Pharmacokinetics and Metabolism (ADME)

The pharmacokinetic profile of Mitiperstat defines its absorption, distribution, metabolism, and excretion (ADME) characteristics in the human body. Studies in healthy volunteers and patient populations have established a consistent profile characterized by rapid absorption, extensive metabolism, and slow, primarily renal, elimination.

3.1 Absorption

Following oral administration, Mitiperstat is rapidly absorbed into the systemic circulation. The time to reach maximum plasma concentration ($T_{max}$) is consistently observed to be between 1 and 2 hours across multiple studies. This rapid absorption profile indicates a quick onset of exposure after dosing. Early clinical studies utilized an oral suspension, while later trials used a tablet formulation; however, bioavailability studies demonstrated similar pharmacokinetic profiles for both formulations, allowing for a seamless transition in development.

3.2 Distribution

Data on the specific tissue distribution of Mitiperstat is limited. However, a human ADME study revealed a key characteristic: the parent drug, Mitiperstat, accounts for only approximately 7% of the total drug-related material circulating in the plasma. This finding indicates that the drug undergoes extensive metabolism, with the majority of the circulating drug-related exposure attributable to its metabolites.

3.3 Metabolism

Mitiperstat is cleared from the body through a combination of metabolic transformation and direct renal excretion. The metabolic pathways are well-characterized:

  • The predominant circulating metabolite is a carbamoyl conjugate known as M7, which accounts for a substantial 75-80% of the total drug-related exposure in plasma. The formation of M7 is primarily catalyzed by the enzyme UDP-glucuronosyltransferase 1A1 (UGT1A1).
  • Another identified metabolic pathway involves N-acetyl transferase 2 (NAT2).
  • Crucially, despite the high systemic concentrations of M7 and other metabolites, they are considered to have little or no inhibitory activity against MPO. This means that the pharmacological effect of the drug is driven almost exclusively by the parent compound, Mitiperstat.

3.4 Elimination

The elimination of Mitiperstat is characterized by an apparent biphasic profile and a notably long duration in the body.

  • Primary Pathway: The main route of elimination for the unchanged parent drug is renal clearance. Studies show that between 32% and 44% of an administered dose is excreted as unchanged Mitiperstat in the urine.
  • Half-Life and Accumulation: Mitiperstat has a long terminal elimination half-life ($t_{1/2}$), reported to be in the range of 38 to 73 hours across various Phase 1 studies. This long half-life supports a convenient once-daily dosing regimen. However, it also leads to drug accumulation with repeated administration. Steady-state concentrations are reached within approximately 10 days of continuous dosing, with an observed accumulation of 2- to 3-fold compared to the first dose.

The pharmacokinetic properties of Mitiperstat have direct implications for its clinical use and safety monitoring. The long half-life and consequent accumulation factor mean that the full therapeutic and potential toxic effects of a given dose are not realized for over a week after initiating therapy. This necessitates careful dose selection and a longer observation period to assess tolerability. Furthermore, in the event of an adverse reaction, the drug will be cleared from the system slowly, extending the duration of the event. This profile is particularly relevant for patients with compromised clearance mechanisms.

3.5 Pharmacokinetics in Special Populations

  • Renal Impairment: Given that renal clearance is the primary elimination pathway for the parent drug, impaired kidney function has a significant impact on Mitiperstat exposure. In a dedicated study, individuals with severe renal impairment (estimated glomerular filtration rate < 30 mL/min) exhibited an approximately two-fold higher total exposure (Area Under the Curve, or AUC) to Mitiperstat compared to matched subjects with normal renal function. This finding is critical for dosing considerations in this vulnerable patient population.
  • Ethnic Differences: Pharmacokinetic studies were conducted in diverse populations. The results showed that the pharmacokinetic profiles of Mitiperstat were similar among healthy Japanese, Chinese, White, and Black/African American volunteers. This consistency indicates that dose adjustments based on ethnicity are not necessary.

Section 4: The Therapeutic Hypothesis: Targeting Myeloperoxidase in Inflammatory Disease

The development of Mitiperstat was founded on a strong scientific rationale targeting the central role of myeloperoxidase in the pathology of chronic inflammatory diseases. This section outlines the biological basis for MPO inhibition as a therapeutic strategy in heart failure and COPD.

4.1 The Role of Myeloperoxidase (MPO) in Pathophysiology

Myeloperoxidase is a heme-containing peroxidase enzyme found in abundance within the azurophilic (primary) granules of neutrophils, a type of white blood cell. Upon activation by inflammatory stimuli, neutrophils release MPO into both the intracellular phagosome and the extracellular space. The primary enzymatic function of MPO is to utilize hydrogen peroxide to catalyze the production of highly reactive chlorinating species, most notably hypochlorous acid (HOCl)—the active component of household bleach.

While this process is a vital component of the innate immune system's microbicidal activity against invading pathogens, the extracellular release of MPO and its reactive products can inflict significant collateral damage on host tissues. This MPO-driven oxidative stress is implicated in a cascade of pathological processes, including endothelial dysfunction, nitric oxide scavenging, lipid peroxidation, and the promotion of interstitial fibrosis. A growing body of evidence has linked elevated plasma levels and activity of MPO to the progression of and poor clinical outcomes in a range of cardiovascular and respiratory diseases.

4.2 Rationale for MPO Inhibition in Heart Failure (HFpEF/HFmrEF)

Heart failure with preserved ejection fraction (HFpEF) is increasingly recognized as a syndrome driven by systemic, comorbidity-induced inflammation. In this paradigm, conditions such as obesity, diabetes, and hypertension create a chronic pro-inflammatory state that leads to coronary microvascular dysfunction, cardiomyocyte stiffness, and interstitial fibrosis—the cardinal features of HFpEF.

The therapeutic hypothesis for Mitiperstat in HFpEF was that MPO is a critical link between systemic inflammation and cardiac pathology. Neutrophils, activated by circulating inflammatory signals, are thought to infiltrate the cardiac microvasculature and interstitium, where they release MPO. The subsequent generation of reactive oxygen species by extracellular MPO was proposed to directly cause the endothelial damage and myocardial remodeling that lead to diastolic dysfunction. Therefore, by selectively inhibiting extracellular MPO, Mitiperstat was hypothesized to break this pathological chain, thereby improving microvascular function, reducing cardiac fibrosis, and ultimately ameliorating the symptoms and exercise intolerance characteristic of HFpEF.

4.3 Rationale for MPO Inhibition in Chronic Obstructive Pulmonary Disease (COPD)

The pathophysiology of COPD is characterized by chronic airway inflammation, with neutrophilic inflammation being the predominant pattern for most patients. In response to long-term irritants like tobacco smoke, massive numbers of neutrophils are recruited to the lungs. These activated neutrophils release their granular contents, including MPO, into the airway lumen and lung tissue.

Observational data strongly support a role for MPO in COPD progression. High levels of MPO in the sputum and serum of COPD patients are directly correlated with accelerated lung function decline, an increased risk of acute exacerbations, and worse cardiovascular outcomes. The hypothesis underpinning the CRESCENDO clinical trial was that MPO-generated oxidants contribute directly to the key features of COPD, including emphysema, small airway fibrosis, and mucus hypersecretion. By administering Mitiperstat, an oral inhibitor with high potency for extracellular MPO, the aim was to reduce lung oxidative stress and inflammation. This was expected to translate into a reduction in the frequency and severity of exacerbations and an improvement in lung function, symptoms, and quality of life for patients with COPD.

Section 5: Clinical Development in Heart Failure

The clinical development program for Mitiperstat in heart failure was designed to first establish proof of mechanism and then to definitively test its efficacy in a large patient population. This journey, from the promising SATELLITE trial to the conclusive ENDEAVOR trial, provides a clear narrative of the drug's performance.

5.1 Phase 2a SATELLITE Trial (NCT03756285): Proof of Mechanism

The SATELLITE trial was a Phase 2a, randomized, double-blind, placebo-controlled study designed as the first test of Mitiperstat in its target patient population. The study enrolled 41 patients with symptomatic heart failure and a left ventricular ejection fraction (LVEF) of 40% or greater (HFpEF or HFmrEF). The primary objectives were to assess the safety of Mitiperstat and, critically, to confirm that it could engage its target in patients.

Key results from the SATELLITE trial were highly encouraging from a pharmacological standpoint:

  • Trial Status: The study was terminated prematurely due to logistical challenges imposed by the COVID-19 pandemic.
  • Target Engagement: The trial successfully met its primary pharmacodynamic endpoint. Treatment with Mitiperstat resulted in a greater than 50% reduction in MPO activity from baseline. When adjusted for the placebo group, this represented a statistically significant 75% decrease in MPO activity, confirming robust target engagement in patients.
  • Safety and Tolerability: Mitiperstat was found to be well tolerated. There were no deaths or treatment-related serious adverse events reported. The most notable treatment-related adverse events were single instances of generalized maculopapular rash, pruritus, and diarrhea.
  • Efficacy Signals: Due to the small sample size and early termination, all efficacy findings were considered exploratory. No statistically significant improvements were observed in secondary endpoints such as coronary flow velocity reserve or 6-minute walk distance. However, investigators noted a positive numerical trend in the Kansas City Cardiomyopathy Questionnaire (KCCQ) overall summary score, a measure of patient-reported health status.

The SATELLITE trial, despite its limitations, was considered a success. It provided the essential proof of mechanism and safety data required to justify moving forward into a larger, more definitive efficacy trial.

5.2 Phase 2b/3 ENDEAVOR Trial (NCT04986202): The Pivotal Efficacy Failure

Building on the results of SATELLITE, AstraZeneca launched the ENDEAVOR trial, a large-scale, international, randomized, double-blind, seamless Phase 2b/3 study designed to definitively evaluate the efficacy and safety of Mitiperstat. The trial enrolled 711 patients with symptomatic HFpEF/HFmrEF (LVEF >40%) across 142 sites in 18 countries. Participants were randomized in a 1:1:1 ratio to receive Mitiperstat 2.5 mg, Mitiperstat 5 mg, or a matching placebo once daily for up to 48 weeks.

The co-primary endpoints were the change from baseline to week 16 in two key measures of patient well-being: the KCCQ Total Symptom Score (KCCQ-TSS) and the 6-Minute Walk Distance (6MWD). The enrolled population was representative of a typical HFpEF cohort, with a mean age of 72 years, 45% female participants, and significant baseline functional limitations (mean 6MWD of 307 meters). Of note, background therapy with sodium-glucose cotransporter 2 (SGLT2) inhibitors, a now-standard treatment for HFpEF, was present in only 21% of patients.

The primary results of the ENDEAVOR trial were unequivocally negative:

  • Primary Efficacy Failure: The trial failed to meet both of its co-primary endpoints. There was no statistically significant difference in the change in KCCQ-TSS or 6MWD at 16 weeks for the pooled Mitiperstat groups compared to placebo.
  • Change in KCCQ-TSS: Treatment difference of -1.4 points (95% CI -3.9 to 1.2; $p=0.29$).
  • Change in 6MWD: Treatment difference of +3.8 meters (95% CI -3.1 to 10.8; $p=0.28$).
  • Secondary Efficacy Results: Consistent with the primary findings, there were no significant improvements in any of the secondary endpoints, which included longer-term assessments of KCCQ and 6MWD, as well as changes in biomarkers (like NT-proBNP) and echocardiographic parameters, at 24 or 48 weeks.
  • Exploratory Cardiovascular Outcomes: An analysis of adjudicated cardiovascular events revealed a numerical trend favoring Mitiperstat, though these findings were not statistically significant and must be interpreted with caution as they were exploratory.
  • Major Adverse Cardiovascular Events (MACE): Hazard Ratio (HR) 0.71 (95% CI 0.42-1.19; $p=0.20$).
  • Heart Failure Hospitalizations: HR 0.64 (95% CI 0.35-1.16; $p=0.14$).

5.3 Synthesis and Interpretation: The Disconnect Between Biology and Clinical Outcomes

The juxtaposition of the SATELLITE and ENDEAVOR trial results provides a clear and compelling narrative. The SATELLITE trial confirmed that Mitiperstat is a potent drug that successfully hits its biological target, MPO, in patients. However, the large, well-powered ENDEAVOR trial demonstrated that hitting this target does not translate into a meaningful clinical benefit for patients in terms of how they feel or function. This outcome critically challenges the foundational therapeutic hypothesis—that systemic MPO inhibition is a sufficient strategy to improve symptoms and functional capacity in a complex, established, and heterogeneous HFpEF population. The link between the target (MPO) and the desired patient-centered outcomes appears to be either non-existent or too weak to be clinically meaningful over a 48-week period.

The non-significant trend toward reduced hospitalizations observed in ENDEAVOR, while numerically intriguing, was ultimately insufficient to alter the drug's fate. The hazard ratios for MACE and HF hospitalizations were based on a relatively small number of events and the high p-values indicate a strong possibility that these findings were due to chance. To confirm such an effect would require a new, even larger, and longer clinical trial focused exclusively on hard outcomes. The investment required for such a trial would be immense. Furthermore, the therapeutic landscape for HFpEF has evolved rapidly, with drug classes like SGLT2 inhibitors and mineralocorticoid receptor antagonists having already demonstrated robust, statistically significant reductions in HF hospitalizations. The bar for entry and commercial viability has been set extremely high. In this context, the weak and exploratory signal from ENDEAVOR was not compelling enough to justify the significant financial risk and resources required for further development, leading to the strategic decision to discontinue the program.

Section 6: Clinical Development in Chronic Obstructive Pulmonary Disease (COPD)

In parallel with its development in heart failure, AstraZeneca investigated Mitiperstat as a potential treatment for Chronic Obstructive Pulmonary Disease (COPD), leveraging the same underlying scientific rationale of targeting neutrophil-driven inflammation and oxidative stress.

6.1 Phase 2a CRESCENDO Trial (NCT05492877): An Innovative Approach

The CRESCENDO trial was a Phase 2a, randomized, double-blind, placebo-controlled study designed to evaluate the efficacy and safety of Mitiperstat in patients with moderate-to-severe COPD who were at a high risk of experiencing exacerbations. The trial planned to enroll approximately 406 participants across roughly 100 global sites.

A key feature of the CRESCENDO trial was its innovative design. Acknowledging the limitations of traditional COPD trials, which often have restrictive inclusion criteria that limit the generalizability of results, CRESCENDO employed a partially decentralized model. This approach was intended to enhance participant recruitment from a broader, more real-world patient population, including through community-based facilities, thereby reducing patient burden and improving the applicability of the findings.

The trial also utilized a novel primary endpoint: the time to the first "CompEx" event. CompEx is a composite endpoint designed to capture a wider spectrum of disease worsening beyond traditional severe exacerbations. It included changes in symptoms, reliever medication use, lung function deterioration, treatment for a moderate or severe exacerbation, or study dropout due to treatment failure. This patient-centric endpoint was designed to be more sensitive in detecting a therapeutic effect.

According to clinical trial registry data, the CRESCENDO trial began in November 2022 and reached its primary and final completion dates in August 2024. However, as of the latest available information, the final results of this study have not been published or presented at a scientific congress.

The innovative design and novel endpoint of the CRESCENDO trial represent a forward-thinking approach to clinical research in COPD. However, the ultimate outcome of the trial and its potential impact remain unknown. The discontinuation of Mitiperstat in its flagship heart failure indication, announced before the completion of CRESCENDO, undoubtedly cast a shadow over the COPD program. Even if the trial were to yield positive results, the overall strategic outlook for the drug at AstraZeneca had already shifted significantly, making the path forward for a new indication highly uncertain. The current absence of published data, despite the trial's completion, represents a critical information gap in the Mitiperstat development story.

Section 7: Comprehensive Safety and Tolerability Assessment

A thorough evaluation of safety and tolerability is paramount in the development of any new therapeutic. Across its entire clinical program, from Phase 1 studies in healthy volunteers to large Phase 2b/3 trials in patients with chronic disease, Mitiperstat was observed to be generally well-tolerated.

7.1 Overall Profile

The safety profile of Mitiperstat was consistent across different populations and study durations. In the large ENDEAVOR trial, the proportion of participants reporting adverse events (AEs) was generally balanced between the Mitiperstat and placebo treatment groups. The most frequently reported AEs (in >5% of participants in any group) were cardiac failure, COVID-19, nasopharyngitis, dizziness, and maculopapular rash.

7.2 Adverse Events of Special Interest

  • Maculopapular Rash: The most distinct and consistent treatment-related adverse event associated with Mitiperstat was a maculopapular (containing both flat and raised lesions) skin rash. This was first noted in a Phase 1 study, where it occurred in participants receiving higher doses of the drug. It was subsequently observed as a treatment-related AE in the Phase 2a SATELLITE trial and was reported in approximately 5% of patients receiving Mitiperstat in the larger ENDEAVOR trial. Importantly, this rash was consistently described as being self-limited, meaning it resolved on its own without requiring major intervention. While not typically severe, this recurring dermatological signal is a key characteristic of the drug's safety profile.

7.3 Cardiac Safety (QT Interval)

Assessing the potential for a new drug to cause cardiac arrhythmias by prolonging the QT interval of the electrocardiogram is a critical regulatory requirement, especially for cardiovascular drugs. AstraZeneca conducted a dedicated "thorough QT" study for Mitiperstat, utilizing advanced concentration-QT (C-QT) modeling with data from a Phase 1 study in healthy volunteers.

The results of this analysis were definitive and reassuring. The model-predicted change in the corrected QT interval (ΔΔQTcF) at the highest anticipated clinical exposure was a mere +0.73 ms. The upper bound of the 90% confidence interval was +3.19 ms, which is well below the 10 ms threshold that triggers regulatory concern. This finding provides strong evidence that Mitiperstat is not associated with a risk of QT interval prolongation at its expected therapeutic doses, effectively ruling out this specific cardiac safety risk.

7.4 Discontinuation due to Adverse Events

In the ENDEAVOR trial, a slightly higher proportion of participants permanently discontinued treatment prematurely in both Mitiperstat groups compared to the placebo group, though specific rates were not detailed in the available synopsis. In the CRESCENDO trial for COPD, adverse events were the most common reason for treatment discontinuation in both the Mitiperstat and placebo arms.

Table 2: Summary of Adverse Events of Special Interest Across Key Trials

TrialAdverse EventMitiperstat Group (Incidence)Placebo Group (Incidence)Comments
SATELLITEMaculopapular Rash1 patient (n=27)0 patients (n=14)Treatment-related, generalized.
SATELLITEPruritus1 patient (n=27)0 patients (n=14)Treatment-related.
SATELLITEDiarrhea1 patient (n=27)0 patients (n=14)Treatment-related.
ENDEAVORMaculopapular Rash~5%<5% (not specified)Described as self-limited; reported in >5% of participants in one of the treatment groups.
ENDEAVORDiscontinuation due to AEsHigher proportion than placeboLower proportion than MitiperstatAEs were a reason for premature treatment discontinuation.

Section 8: Program Discontinuation, Regulatory Status, and Final Analysis

The culmination of Mitiperstat's development journey was not regulatory approval, but a strategic discontinuation based on the totality of the clinical evidence. This final section addresses the ultimate fate of the program and provides an expert conclusion on its legacy.

8.1 Development Status and Discontinuation

Mitiperstat successfully advanced through early and mid-stage clinical development, reaching a seamless Phase 2b/3 trial, its highest stage of development. However, following the unblinding of the ENDEAVOR trial results, the program's trajectory changed.

In its Q4 2024 earnings report in early 2025, AstraZeneca formally announced the discontinuation of Phase 2 trials of Mitiperstat for both heart failure and metabolic dysfunction-associated steatohepatitis (NASH). The company's official rationale for this decision was "strategic portfolio prioritization". This phrasing is common in the pharmaceutical industry and typically signifies that a drug's clinical profile does not meet the internal criteria for further investment, especially when compared to other assets in the company's pipeline and the external competitive landscape.

8.2 Regulatory Status

Mitiperstat remains an investigational compound. It has not received marketing authorization from any major regulatory body, including the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA). Its legal status is "Investigational". As it has never been approved for commercial use, there are no official drug label annotations from regulatory agencies detailing pharmacogenomic or other specific information. The drug has been studied in clinical trials under Investigational New Drug (IND) applications, but it has not progressed to a New Drug Application (NDA) or Marketing Authorisation Application (MAA) submission.

8.3 Final Analysis and Expert Conclusion

The Mitiperstat program represents a case of exemplary pharmaceutical science meeting the harsh realities of clinical translation. The drug itself is a product of sophisticated medicinal chemistry—a potent, selective, and irreversible inhibitor designed with precision against a biologically plausible target. The early clinical program was executed effectively, successfully demonstrating that Mitiperstat could robustly and safely inhibit MPO activity in humans, thereby achieving its pharmacological goal.

However, the program's ultimate failure underscores a critical lesson in drug development: a compelling mechanism and successful target engagement do not guarantee clinical efficacy. The large, well-conducted ENDEAVOR trial provided a definitive answer to the key clinical question, and that answer was negative. The inability of Mitiperstat to improve how patients with HFpEF feel or function demonstrated a fundamental disconnect between the therapeutic hypothesis and the patient experience.

The official reason for termination, "strategic portfolio prioritization," should be interpreted as a direct business consequence of the negative ENDEAVOR data. The weak, non-significant, and exploratory signal on reducing hospitalizations was simply not a strong enough foundation upon which to build another multi-year, multi-hundred-million-dollar confirmatory outcomes trial. This is particularly true in the context of the highly competitive HFpEF market, where therapies like SGLT2 inhibitors have already established a high standard of care with proven benefits.

While the final results of the CRESCENDO trial in COPD are not yet public, the discontinuation of the flagship indications makes the future of Mitiperstat as a viable therapeutic agent highly improbable. Although the program did not result in a new medicine, it has made a valuable contribution to the field. It has provided high-quality evidence suggesting that systemic inhibition of myeloperoxidase, at least with this agent and in this patient population, is not an effective strategy for managing the symptomatic burden of HFpEF. This "negative" data is crucial for guiding future research efforts toward more promising pathways.

Works cited

  1. Mitiperstat - Wikipedia, accessed October 24, 2025, https://en.wikipedia.org/wiki/Mitiperstat
  2. Mitiperstat - AstraZeneca - AdisInsight - Springer, accessed October 24, 2025, https://adisinsight.springer.com/drugs/800044998
  3. Mitiperstat (AZD4831) | MPO Inhibitor - MedchemExpress.com, accessed October 24, 2025, https://www.medchemexpress.com/mitiperstat.html
  4. Pharmacokinetics and Tolerability of the Novel Myeloperoxidase ..., accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11564280/
  5. Rationale and design of ENDEAVOR: A sequential phase 2b-3 randomized clinical trial to evaluate the effect of myeloperoxidase inhibition on symptoms and exercise capacity in heart failure with preserved or mildly reduced ejection fraction - PubMed, accessed October 24, 2025, https://pubmed.ncbi.nlm.nih.gov/37470101/
  6. The myeloperoxidase inhibitor mitiperstat (AZD4831) does not prolong the QT interval at expected therapeutic doses - NIH, accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10915730/
  7. Myeloperoxidase Inhibition in Heart Failure With Preserved or Mildly Reduced Ejection Fraction: SATELLITE Trial Results - PubMed, accessed October 24, 2025, https://pubmed.ncbi.nlm.nih.gov/37072105/
  8. AstraZeneca's investigational MPO inhibitor AZD4831 shows promise in patients with heart failure and a preserved ejection fraction, accessed October 24, 2025, https://www.astrazeneca.com/media-centre/medical-releases/astrazenecas-investigational-mpo-inhibitor-azd4831-shows-promise-in-patients-with-heart-failure-and-a-preserved-ejection-fraction.html
  9. Myeloperoxidase Inhibition With Mitiperstat in Heart Failure With Preserved and Mildly Reduced Ejection Fraction - ENDEAVOR - American College of Cardiology, accessed October 24, 2025, https://www.acc.org/latest-in-cardiology/clinical-trials/2024/11/17/16/35/endeavor
  10. ENDEAVOR: Mitiperstat vs. Placebo in Patients With HFpEF or ..., accessed October 24, 2025, https://www.acc.org/latest-in-cardiology/articles/2024/11/13/21/17/mon-9am-endeavor-aha-2024
  11. (PDF) The myeloperoxidase inhibitor mitiperstat ( AZD4831 ) does not prolong the QT interval at expected therapeutic doses - ResearchGate, accessed October 24, 2025, https://www.researchgate.net/publication/378783987_The_myeloperoxidase_inhibitor_mitiperstat_AZD4831_does_not_prolong_the_QT_interval_at_expected_therapeutic_doses
  12. The myeloperoxidase inhibitor mitiperstat (AZD4831) does not prolong the QT interval at expected therapeutic doses - PubMed, accessed October 24, 2025, https://pubmed.ncbi.nlm.nih.gov/38445541/
  13. AstraZeneca CEO defends 'fantastic' Alexion buy as halts drug, accessed October 24, 2025, https://www.fiercebiotech.com/biotech/astrazeneca-resets-big-bets-taking-1b-charges-ex-alexion-fusion-assets-suffer-setbacks
  14. An Efficacy and Safety Study of Mitiperstat (AZD4831) (MPO inhibitor) vs placebo in the treatment of moderate to severe COPD. - CRESCENDO - AstraZeneca Clinical Trials, accessed October 24, 2025, https://www.astrazenecaclinicaltrials.com/study/D6582C00001/
  15. (PDF) Phase 2a, Randomized Trial of Mitiperstat Versus Placebo in Patients with COPD at High Risk of Exacerbation (CRESCENDO) - ResearchGate, accessed October 24, 2025, https://www.researchgate.net/publication/395826504_Phase_2a_Randomized_Trial_of_Mitiperstat_Versus_Placebo_in_Patients_with_COPD_at_High_Risk_of_Exacerbation_CRESCENDO
  16. NCT05492877 | An Efficacy and Safety Study of Mitiperstat (AZD4831) (MPO Inhibitor) vs Placebo in the Treatment of Moderate to Severe COPD. | ClinicalTrials.gov, accessed October 24, 2025, https://clinicaltrials.gov/study/NCT05492877
  17. Mitiperstat - Drug Targets, Indications, Patents - Patsnap Synapse, accessed October 24, 2025, https://synapse.patsnap.com/drug/aca3a16421854c9cb257418c0cf00264
  18. Mitiperstat | C15H15ClN4OS | CID 121362450 - PubChem - NIH, accessed October 24, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Mitiperstat
  19. pmc.ncbi.nlm.nih.gov, accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11564280/#:~:text=Mitiperstat%20(AZD4831)%20is%20a%20mechanism,the%20enzymatic%20activity%20%5B5%5D.
  20. Pharmacokinetics and Tolerability of the Novel Myeloperoxidase Inhibitor Mitiperstat in Healthy Japanese and Chinese Volunteers - PubMed, accessed October 24, 2025, https://pubmed.ncbi.nlm.nih.gov/39495467/
  21. Phase 2a, Randomized Trial of Mitiperstat Versus Placebo in Patients with COPD at High Risk of Exacerbation (CRESCENDO) - PubMed Central, accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12477066/
  22. Rationale and design of ENDEAVOR: a sequential phase 2b–3 randomized clinical trial to evaluate the effect of myeloperoxidase inhibition on symptoms and exercise capacity in heart failure with preserved or mildly reduced ejection fraction, accessed October 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10592288/
  23. NCT04986202 | Study to Evaluate the Efficacy and Safety of AZD4831 in Participants With Heart Failure With Left Ventricular Ejection Fraction > 40% | ClinicalTrials.gov, accessed October 24, 2025, https://clinicaltrials.gov/study/NCT04986202
  24. Late-Breaking Science Abstracts and Featured Science Abstracts From the American Heart Association's Scientific Sessions 2024 and Late-Breaking Abstracts in Resuscitation Science From the Resuscitation Science Symposium 2024 | Circulation - American Heart Association Journals, accessed October 24, 2025, https://www.ahajournals.org/doi/10.1161/CIR.0000000000001299
  25. COPD clinical trial of mitiperstat now recruiting adults in Oklahoma, accessed October 24, 2025, https://copdnewstoday.com/news/copd-clinical-trial-mitiperstat-now-recruiting-adults-oklahoma/
  26. A Phase IIa Randomised, Double Blind, Placebo Controlled, Parallel Arm, Multi- Centre Study to Evaluate the Efficacy and - Onderzoek met mensen, accessed October 24, 2025, https://onderzoekmetmensen.nl/en/node/53515/pdf
  27. mitiperstat (AZD4831) News - LARVOL Sigma, accessed October 24, 2025, https://sigma.larvol.com/product.php?e1=732933&tab=newstrac
  28. Redacted for Public Disclosure - Amazon S3, accessed October 24, 2025, https://s3.amazonaws.com/ctr-med-7111/D6580C00010/ac832783-a7d5-46b3-8823-8d5a77d1fa45/8d759732-63d6-43eb-91e9-9c058b07a8e1/D6580C00010-Redacted_CSR_synopsis-v2.pdf
  29. A Phase IIa Randomised, Double Blind, Placebo Controlled, Parallel Arm, Multi-Centre Study to Evaluate the Efficacy and Safety of Mitiperstat (AZD4831), for 12-24 Weeks, in Patients with Moderate to Severe Chronic Obstructive Pulmonary Disease (COPD) | Research with human participants, accessed October 24, 2025, https://onderzoekmetmensen.nl/en/trial/53515
  30. Drug Label Annotations - mitiperstat - ClinPGx, accessed October 24, 2025, https://www.clinpgx.org/drug/PA166358101/labelAnnotation

Published at: October 24, 2025

This report is continuously updated as new research emerges.

MedPath

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

© 2025 MedPath, Inc. All rights reserved.