Small Molecule
C26H28N6O
1254036-71-9
Nemiralisib (GSK2269557) is an investigational small molecule drug developed by GlaxoSmithKline (GSK) as a potent and highly selective inhibitor of phosphoinositide 3-kinase delta (PI3Kδ).[1] Designed for inhaled administration, its primary therapeutic objective was the treatment of inflammatory respiratory diseases, with a principal focus on Chronic Obstructive Pulmonary Disease (COPD) and a secondary, mechanism-based exploration in the rare primary immunodeficiency, Activated PI3Kδ Syndrome (APDS).[1] The biological rationale for targeting PI3Kδ was robust, supported by evidence of pathway upregulation in COPD and direct genetic validation from APDS, a condition caused by gain-of-function mutations in the target enzyme.[3]
Early-phase clinical development was promising, demonstrating that inhaled nemiralisib could successfully engage its target in the human lung, leading to a dose-dependent reduction in key biomarkers of PI3Kδ activity.[5] However, this biochemical success failed to translate into clinical benefit. The development program was ultimately halted following the termination of a large, dose-ranging Phase IIb clinical trial (NCT03345407) in patients with acute exacerbations of COPD.[6] An interim futility analysis of this study revealed a comprehensive lack of efficacy across all doses tested, with no improvement in the primary endpoint of lung function (FEV1) or in key secondary endpoints such as the rate of re-exacerbations.[4] Furthermore, the drug exhibited a challenging safety profile at higher doses, characterized by a dose-dependent increase in post-inhalation cough.[4] Parallel investigations in APDS also failed to demonstrate target engagement or clinical efficacy, suggesting a fundamental issue with the therapeutic approach or delivery modality in diseased lungs.[3]
The discontinuation of nemiralisib was driven by this clear clinical failure, which occurred concurrently with a major strategic R&D reprioritization at GSK. During the 2017-2019 period, the company shifted its focus away from respiratory diseases towards oncology and immunology, leading to the culling of numerous pipeline assets, including nemiralisib.[6] The confluence of definitive negative clinical data and this evolving corporate strategy sealed the fate of the program. This report provides an exhaustive analysis of nemiralisib, from its molecular characteristics and pharmacological profile to a detailed deconstruction of its clinical trial results and the dual factors that precipitated its discontinuation.
To establish the precise identity of the molecule, a comprehensive list of its nomenclature and registry identifiers is essential.
Nemiralisib is a new molecular entity classified as a small molecule drug.[1] Its structure incorporates several heterocyclic ring systems, placing it within multiple chemical classes, including indazoles, indoles, oxazoles, piperazines, and pyrazoles.[1]
| Property | Value | Source | 
|---|---|---|
| Water Solubility | 0.0571 mg/mL | ALOGPS 10 | 
| logP (Lipophilicity) | 3.79 | ALOGPS 10 | 
| pKa (Strongest Acidic) | 11.32 | Chemaxon 10 | 
| pKa (Strongest Basic) | 7.82 | Chemaxon 10 | 
| Polar Surface Area | 76.98 $Å^2$ | Chemaxon 10 | 
| Lipinski's Rule of Five | Yes | Chemaxon 10 | 
| Bioavailability (Predicted) | 1 | Chemaxon 10 | 
The very low water solubility is a significant characteristic for an inhaled drug, as dissolution in the lung's epithelial lining fluid is a prerequisite for target engagement. Conversely, its compliance with Lipinski's Rule of Five suggests properties generally favorable for membrane permeation and absorption.[10]
Nemiralisib was developed in several forms to optimize its delivery and stability.
The deliberate effort to improve the delivery formulation from the Diskus to the Ellipta device underscores an acknowledgment of the challenges posed by the drug's physicochemical properties, particularly its poor water solubility. Enhancing the fine particle fraction was a logical and necessary step to maximize the amount of drug reaching the target tissue. However, the ultimate failure of the large-scale COPD trial, which utilized the optimized delivery system, indicates that improved formulation alone was insufficient to elicit a clinical response. This outcome suggests that the therapeutic hypothesis was flawed or that even with enhanced delivery, therapeutically relevant concentrations could not be achieved or sustained within the complex, inflamed microenvironment of a diseased lung. This latter possibility is echoed in the findings from the APDS trial, where investigators speculated that the drug was not retained in structurally damaged lungs for a sufficient duration.[3]
The rationale for developing nemiralisib is rooted in the specific biological role of its target, the delta isoform of phosphoinositide 3-kinase (PI3Kδ).
Medicinal chemistry efforts for nemiralisib resulted in a molecule with a highly optimized profile for its intended target.
A crucial step in the early clinical development of any targeted therapy is to confirm that the drug can reach its target in humans and exert the expected biological effect. Nemiralisib successfully cleared this hurdle.
The clinical development of nemiralisib presents a stark example of the translational chasm that frequently plagues drug development. From a molecular and early clinical perspective, the drug was a success. It was a potent and exquisitely selective molecule designed against a well-validated biological target.[2] The early human studies provided clear and unambiguous evidence of in vivo target engagement, demonstrating a reduction in the key downstream biomarker, $PIP_3$, in the target organ.[5] This successful outcome at the biochemical level, however, failed to translate into any meaningful clinical benefit in large-scale patient trials for either COPD or APDS.[3] This disconnect suggests that the initial therapeutic hypothesis—that inhibiting the PI3Kδ pathway and reducing $PIP_3$ levels in the lung would be sufficient to meaningfully alter the complex, multifactorial pathophysiology of a COPD exacerbation—was ultimately incorrect. The link between this proximal biomarker modulation and the desired clinical outcomes of improved lung function and reduced exacerbations was not as robust as anticipated. It is plausible that in the context of established COPD, the PI3Kδ pathway is either a less dominant driver of clinical symptoms than other inflammatory pathways or that its inhibition is insufficient to overcome redundant signaling mechanisms that sustain the disease process.
The pharmacokinetic profile of nemiralisib was characterized through a series of Phase I studies in healthy volunteers, employing various administration routes to build a comprehensive understanding of its absorption, distribution, metabolism, and excretion (ADME) properties.
Once absorbed, nemiralisib distributed throughout the body, with a notable preference for the target organ.
The clearance of nemiralisib from the body is primarily driven by hepatic metabolism.
The pharmacokinetic profile of nemiralisib, particularly its long half-life, can be viewed as a double-edged sword. On one hand, the ~40-hour half-life and predictable accumulation were advantageous, strongly supporting a convenient once-daily dosing regimen, which is highly desirable for patient adherence in chronic diseases like COPD.[5] On the other hand, this same property presents a potential liability. The dose-ranging COPD trial revealed a clear signal for dose-dependent adverse events, most notably post-inhalation cough, which became significantly more prevalent at the highest doses tested.[4] For a drug with a long half-life, the onset of such an adverse event means that it will take a considerable amount of time for the drug to clear from the system, potentially prolonging the patient's discomfort or risk. This accumulation, coupled with the evidence of systemic distribution, increases the potential for both local and systemic side effects. This dynamic may have created a significant therapeutic window challenge: the doses required to potentially achieve efficacy may have pushed the accumulating drug concentrations into a range that caused unacceptable local tolerability issues, thereby narrowing or eliminating the path to a successful dose. The ultimate failure to identify an effective and well-tolerated dose in the comprehensive Phase IIb study supports this interpretation.[8]
The clinical development of nemiralisib followed a logical, phased approach, progressing from initial human safety and pharmacokinetic studies to larger trials designed to evaluate efficacy in specific patient populations. The program was comprehensive but ultimately unsuccessful, with key trials being terminated or failing to meet their objectives, leading to the discontinuation of the drug's development. A summary of the key clinical trials is presented in the table below.
| Trial ID | Phase | Status | Condition(s) | Purpose | Key Findings / Outcome | Source Snippets | 
|---|---|---|---|---|---|---|
| NCT03345407 | 2 | Terminated | Chronic Obstructive Pulmonary Disease (COPD) | Treatment, Dose-Finding | Failed to meet primary endpoint (FEV1); no improvement in re-exacerbations. Development discontinued based on futility analysis. | 4 | 
| NCT02593539 | 2 | Completed | Activated PI3K-delta Syndrome (APDS) | Treatment | Safe and well-tolerated, but no convincing evidence of target engagement or clinical efficacy. Development for APDS suspended. | 3 | 
| NCT03398421 | 1 | Completed | Healthy Subjects | Drug-Drug Interaction | Assessed the pharmacokinetic effect of the potent CYP3A4 inhibitor itraconazole on nemiralisib. Results were posted. | 6 | 
| NCT03315559 | 1 | Completed | Healthy Subjects | ADME | Determined the absorption, distribution, metabolism, and excretion profile using a radiolabelled microtracer dose. | 6 | 
| NCT03189589 | 1 | Completed | Healthy Subjects | PK / Formulation | Assessed the pharmacokinetics and safety of a new dry powder formulation in the Ellipta inhaler, supporting its use in Phase IIb. | 15 | 
| NCT02691325 | 1 | Completed | Healthy Subjects | PK / Bioavailability | Evaluated single and repeat doses via the Ellipta DPI and quantified the oral contribution to systemic exposure using a charcoal block. | 22 | 
The development trajectory began with a series of foundational Phase I studies in healthy volunteers. These trials successfully established the drug's safety profile at various doses, characterized its pharmacokinetic properties (including its long half-life and accumulation), evaluated the performance of new inhaler formulations, and investigated its metabolic pathways and potential for drug-drug interactions.[15] With this supportive Phase I data package, GSK advanced nemiralisib into two parallel Phase II programs to test its efficacy in patients: a large, pivotal study in COPD and a smaller, exploratory study in the rare disease APDS.[1] It was at this critical efficacy-testing stage that the program faltered, as neither trial was able to demonstrate a clinical benefit, leading to the cessation of all development activities.[3]
The cornerstone of the nemiralisib development program for COPD was the NCT03345407 study, a large-scale, multicenter trial designed to definitively assess the drug's dose-response relationship, efficacy, and safety.[6] The study was a Phase IIb, randomized, double-blind, placebo-controlled, parallel-group trial that enrolled 938 patients (aged 40-80 years with a significant smoking history) who were experiencing an acute moderate or severe exacerbation of COPD requiring standard-of-care treatment.[4]
The rationale was to intervene during a period of heightened inflammation to see if nemiralisib could improve recovery and prevent subsequent events. Patients were randomized to receive either placebo or one of six active doses of inhaled nemiralisib (12.5 µg, 50 µg, 100 µg, 250 µg, 500 µg, or 750 µg) administered once daily for a 12-week treatment period, followed by a 12-week follow-up period. All treatment was given in addition to the patient's standard-of-care therapy.[4]
The trial failed to demonstrate any clinical benefit for nemiralisib across all doses tested. The results represented a comprehensive efficacy failure.
While nemiralisib failed on efficacy, it also presented a challenging tolerability profile, particularly at higher doses.
Given the emerging data, GSK made the decision to halt the study prematurely. Recruitment was stopped after a planned interim futility analysis was conducted. The results of this analysis indicated that there was a very low probability of the study meeting its primary success criteria if it were to continue to completion.[4] This definitive clinical data served as the primary trigger for the termination of the NCT03345407 trial and, by extension, the entire nemiralisib program for COPD.
The results from this large, well-designed, dose-ranging study revealed what can be described as an inverted therapeutic window. In a successful drug development program, as the dose increases, efficacy should rise before the incidence of dose-limiting toxicities becomes unacceptable. For nemiralisib in COPD, the opposite occurred. The efficacy curve across a wide range of doses was flat and indistinguishable from placebo.[4] Concurrently, the toxicity curve, driven primarily by dose-dependent cough, was clearly rising.[4] This created a situation where increasing the dose worsened tolerability without providing any corresponding clinical benefit. Such a profile is unviable for a therapeutic agent, making the decision to terminate the program based on the futility analysis a scientifically and commercially sound one.
In parallel with the large COPD program, GSK initiated a smaller, more targeted study of nemiralisib in patients with Activated PI3Kδ Syndrome (APDS). APDS is a rare, primary inborn error of immunity caused by autosomal dominant gain-of-function mutations in one of the two genes encoding the PI3Kδ enzyme ($PIK3CD$ or $PIK3R1$).[3] This genetic defect leads to constitutive activation of the PI3Kδ pathway in immune cells, resulting in a complex clinical phenotype characterized by recurrent sinopulmonary infections, bronchiectasis, lymphoproliferation, and immunodeficiency.[3]
The rationale for studying nemiralisib in this population was exceptionally strong. It represented a precision medicine approach, where the drug's specific mechanism of action—the inhibition of PI3Kδ—directly counteracts the underlying molecular pathology of the disease. This provided a "best-case scenario" to demonstrate the drug's biological activity in a human disease context.
The investigation was conducted as a small, open-label trial (NCT02593539). The study enrolled five subjects with a confirmed diagnosis of APDS, who were treated with inhaled nemiralisib for a period of 12 weeks. The primary objectives were to evaluate the safety, systemic exposure, and effects on lung and systemic biomarkers of PI3Kδ pathway activity.[3]
The trial demonstrated that nemiralisib was generally safe in this small patient population but failed to show any evidence of biological activity or clinical benefit.
Based on the complete lack of evidence for target engagement or downstream efficacy, the study's authors concluded that the data did not support the hypothesis that inhaled nemiralisib would benefit patients with APDS. Consequently, the clinical development of nemiralisib for this indication was suspended.[3]
The failure of nemiralisib in APDS is particularly illuminating and presents a significant paradox. This was the indication with the most direct and powerful biological rationale, where the drug's mechanism was perfectly matched to the disease's genetic cause. The failure in this context points to a problem more fundamental than a simple mechanism-disease mismatch, as might be argued for a complex, heterogeneous disease like COPD. The investigators' hypothesis provides a compelling explanation: the pathophysiology of the target organ itself actively undermined the drug's intended pharmacokinetic profile. The structural lung damage caused by the disease may have prevented the inhaled drug from being retained locally for a sufficient duration to engage its target effectively. This creates a challenging paradox for drug development: the very disease that makes a patient an ideal candidate for a drug's mechanism of action can simultaneously make them a poor candidate for its route of administration. This experience serves as a critical lesson for the development of inhaled therapies for any disease that causes significant structural lung damage, suggesting that a systemic (e.g., oral) route of administration might be necessary even when the primary target organ is the lung.
The primary and most direct cause for the discontinuation of the nemiralisib development program was its definitive failure to demonstrate clinical efficacy. This conclusion is not based on ambiguous signals or marginal results but on the unambiguous negative outcome of a large, well-designed, and rigorously conducted Phase IIb trial in its lead indication, COPD. The decision by GSK to halt trial NCT03345407 early, based on a pre-planned interim futility analysis, underscores the clarity of the negative data.[4] The failure was comprehensive, with no benefit observed across the primary endpoint of lung function, the key secondary endpoint of exacerbation reduction, or in any patient-reported outcomes, across a wide spectrum of six different doses.[4] This lack of a dose-response for efficacy, coupled with a clear dose-response for the key adverse event of cough, painted a clear picture of an unviable therapeutic profile.[4]
This clinical verdict was further solidified by the disappointing results from the APDS trial. The failure to demonstrate target engagement or biomarker modulation in this "best-case" patient population—where the drug's mechanism directly opposed the disease's genetic driver—suggested that the issues with nemiralisib were fundamental, relating either to the therapeutic hypothesis or, more likely, the viability of an inhaled delivery approach in structurally compromised lungs.[3]
The clinical failure of nemiralisib did not occur in a corporate vacuum. Its demise coincided with a period of significant strategic transformation within GSK's pharmaceutical R&D division. Beginning in 2017, under the new leadership of CEO Emma Walmsley, the company initiated a major pipeline overhaul designed to improve R&D productivity and focus investment on areas with the highest potential for growth and scientific innovation.[29] This strategy involved culling a large number of preclinical and clinical programs—over 80 by early 2019—and concentrating 80% of the R&D budget into four priority therapeutic areas: HIV/infectious diseases, oncology, and immuno-inflammation, with respiratory being a notable area of de-emphasis.[9]
In a February 2019 update on this R&D restructuring, GSK explicitly named nemiralisib as one of the assets being discontinued. It was part of a "cull" of respiratory medicines that also included another Phase II COPD drug, danirixin. This move was framed as part of a deliberate redirection of the R&D engine away from its former heartland of respiratory medicine and into the targeted new growth areas.[6]
The discontinuation of nemiralisib is best understood as the result of a powerful confluence of these two factors: unequivocal clinical failure and a concurrent, top-down corporate strategic shift. The negative data from the NCT03345407 futility analysis provided the clear, data-driven, scientific rationale to terminate the program. There was no viable path forward from a clinical or regulatory perspective. Simultaneously, the drug's status as a respiratory asset made it a prime candidate for discontinuation under GSK's new R&D strategy, which was actively de-prioritizing the therapeutic area.
This intersection of events created what might be termed a "low bar for failure." For a drug candidate in a non-priority therapeutic area, any significant negative data is highly likely to be a terminal event, prompting a swift decision to cut losses and reallocate resources. In contrast, a similar clinical setback for an asset in a high-priority area, such as oncology, might have triggered additional investment in biomarker analysis, subgroup identification, or exploration of combination therapies in an attempt to salvage the program. For nemiralisib, the clinical failure was clear, but the strategic context ensured that the decision to terminate would be swift, decisive, and final. Its story is therefore not just a case study in clinical trial failure but also a clear example of how modern pharmaceutical portfolio management operates, where go/no-go decisions are a function of both scientific data and the dynamic strategic priorities of the parent organization.
Published at: October 27, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.