MedPath

Namilumab Advanced Drug Monograph

Published:Jun 12, 2025

Generic Name

Namilumab

Drug Type

Biotech

CAS Number

1206681-39-1

Namilumab (DB13037): A Comprehensive Clinical and Pharmacological Review

I. Executive Summary

Namilumab is an investigational human IgG1 kappa monoclonal antibody designed to target and neutralize granulocyte-macrophage colony-stimulating factor (GM-CSF), a cytokine implicated in various inflammatory and autoimmune diseases. Its therapeutic rationale centers on mitigating the pro-inflammatory effects of GM-CSF. Namilumab has been evaluated in clinical trials for several conditions, including Rheumatoid Arthritis (RA), Plaque Psoriasis, Axial Spondyloarthritis (AS), Sarcoidosis, and COVID-19 pneumonia.[1] Despite a strong biological basis for targeting GM-CSF, Namilumab's clinical development has been marked by a general trend of discontinuation for chronic inflammatory diseases, primarily due to insufficient efficacy. This pattern suggests that while GM-CSF is involved in these conditions, its role may not be universally dominant or easily modulated to achieve broad therapeutic success. In contrast, some positive signals emerged in the context of acute inflammation, such as in COVID-19 pneumonia, although further development in this area has not been actively pursued based on available information.[4] The drug's journey through multiple pharmaceutical companies highlights the inherent challenges and high attrition rates in drug development, even for molecules with well-characterized mechanisms and targets, where strategic business decisions often intersect with scientific findings to shape an asset's trajectory.[4]

II. Namilumab: Drug Profile and Mechanism of Action

A. Key Identifiers and Physicochemical Properties

Namilumab, also known by the alternative identifiers MT203 and AMG203, is a biotech product classified as a protein-based therapy, specifically a human Immunoglobulin G1 (IgG1) kappa monoclonal antibody.[1] Its DrugBank Accession Number is DB13037, and its CAS Number is 1206681-39-1.[1] The sequence origin of the antibody is human, and it has an approximate molecular weight of 145.78 kDa.[8] The selection of a human IgG1 kappa isotype for Namilumab aligns with established practices for therapeutic antibodies targeting soluble cytokines, aiming to optimize efficacy and half-life while minimizing the potential for immunogenic responses in patients.[8]

Table 1: Namilumab - Key Drug Identifiers and Properties

PropertyDetailSnippet References
Generic NameNamilumab1
Other Names/IdentifiersMT203, AMG2031
DrugBank IDDB130371
CAS Number1206681-39-19
TypeBiotech1
Biologic ClassificationHuman IgG1 kappa Monoclonal Antibody8
Pharmacological TargetGranulocyte-Macrophage Colony-Stimulating Factor (GM-CSF / CSF2)1
Approx. Molecular Weight145.78 kDa9
Sequence OriginHuman8

B. Pharmacological Target: Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF)

Namilumab targets granulocyte-macrophage colony-stimulating factor (GM-CSF), also known as colony-stimulating factor 2 (CSF2).[1] GM-CSF is a well-recognized pro-inflammatory cytokine crucial for the proliferation, differentiation, survival, and activation of myeloid cells, including granulocytes (neutrophils, eosinophils, basophils) and macrophages.[2] It stimulates these cells to produce other inflammatory mediators, such as TNF-α, IL-1, and IL-6, thereby amplifying inflammatory responses.[11] In various autoimmune and chronic inflammatory conditions, GM-CSF is found to be overproduced, with elevated levels detected in inflamed tissues, such as the synovial fluid of patients with rheumatoid arthritis, and implicated in the pathogenesis of diseases like psoriasis and axial spondyloarthritis.[2] Its role as a key activator of the innate immune system positions it as a logical target for therapeutic intervention in these disorders.[11]

C. Mechanism of Action

Namilumab exerts its therapeutic effect by specifically binding with high affinity to the GM-CSF ligand.[1] This binding neutralizes the biological activity of GM-CSF, preventing it from interacting with its cell surface receptors on target myeloid cells.[2] By blocking GM-CSF signaling, Namilumab aims to reduce the recruitment, activation, and survival of inflammatory cells at sites of inflammation, thereby diminishing the production of downstream inflammatory mediators and potentially alleviating disease symptoms.[2] The GM-CSF receptor signaling cascade involves the phosphorylation of STAT5 by Janus kinase 2 (JAK2); Namilumab, by sequestering GM-CSF, indirectly modulates this downstream pathway.[10] The overarching goal of this mechanism is to reduce inflammation and modulate the immune response in conditions where GM-CSF plays a significant pathological role.[3] While GM-CSF is a clearly defined pro-inflammatory cytokine, the clinical trial outcomes for Namilumab suggest that its role as an indispensable driver may be less universal across different chronic inflammatory conditions than initially hypothesized, or that the level of neutralization achieved was insufficient in specific disease contexts.

III. Clinical Development Program and Efficacy

Namilumab has undergone an extensive clinical development program, investigating its efficacy and safety across a range of inflammatory and autoimmune diseases. These include Rheumatoid Arthritis, Plaque Psoriasis, Axial Spondyloarthritis, Sarcoidosis, and more recently, COVID-19 pneumonia. The outcomes of these trials have been varied, with notable successes in early-phase RA studies and some positive signals in acute COVID-19 pneumonia, but general failure to meet primary endpoints in larger studies for chronic conditions, leading to discontinuation for most of these indications.

Table 2: Summary of Key Namilumab Clinical Trials

IndicationTrial ID / NamePhaseKey Efficacy Endpoint(s)Summary of Efficacy ResultsKey Safety ObservationsDevelopment StatusSnippet References
Rheumatoid Arthritis (mild-moderate)NCT01317797 / PRIORAIbSafety/Tolerability; DAS28-CRP changeGreater improvement in DAS28-CRP, SJC, TJC vs placebo (post-hoc). Significant DAS28-CRP improvement at Day 43 & 99.Generally well tolerated; TEAEs similar to placebo (nasopharyngitis, RA exacerbation). No ADAs.Completed11
Rheumatoid Arthritis (moderate-severe)NCT02393378 / EUCTR2013-002805-76-GB (NEXUS)IIChange in DAS28-CRP at week 12Significant difference for 150mg vs placebo (p=0.005). Higher ACR50 for 150mg.Generally well tolerated. 1 SAE (MI) in 150mg group. No serious infections.Completed, but indication later discontinued14
Plaque Psoriasis (moderate-severe)NCT02129777 / NEPTUNEIIPASI 75 at week 12Failed to meet primary endpoint. Low PASI 75 response in all groups, no significant difference vs placebo.Tolerability consistent with other studies.Discontinued due to lack of efficacy9
Axial Spondyloarthritis (Ankylosing Spondylitis)NCT03622658 / NAMASTEIIaASAS20 at week 12Failed to meet primary endpoint. Lower ASAS20 in namilumab group vs placebo.Generally well tolerated; TEAE rates similar to placebo.Completed, failed to show efficacy6
Pulmonary Sarcoidosis (chronic active)NCT05314517 / RESOLVE-LungIIProportion of subjects with a Rescue EventFailed to meet primary endpoint. No consistent benefit in secondary endpoints.Safety profile consistent with previous studies.Discontinued due to lack of efficacy (Dec 2024)9
Cardiac SarcoidosisNCT05351554 / RESOLVE-HeartIIN/A (terminated early)N/AN/ATerminated due to sponsor business decision (2022)7
COVID-19 Pneumonia (hospitalized, high CRP)CATALYST (ISRCTN40580903)IICRP reduction over time97% probability of CRP reduction vs usual care. Fewer deaths, more discharges (trends).Generally well tolerated, similar AE rates to usual care.Completed, recommended for further study, but "no development reported" later5

A. Rheumatoid Arthritis (RA)

Namilumab's investigation in Rheumatoid Arthritis began with promising early-phase results. The Phase 1b PRIORA study (NCT01317797) evaluated three subcutaneous (SC) injections of namilumab (150 mg or 300 mg) or placebo on days 1, 15, and 29 in patients with active, mild-to-moderate RA on stable methotrexate doses.[11] The primary outcome was safety and tolerability. Namilumab was generally well tolerated, with treatment-emergent adverse events (TEAEs) like nasopharyngitis and RA exacerbation occurring at similar rates across groups. Importantly, no anti-namilumab antibodies were detected, and its pharmacokinetics were linear and typical for an SC mAb. A post-hoc efficacy analysis (per protocol, n=21) indicated greater improvements in Disease Activity Score 28 (DAS28) using both erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP), swollen joint counts (SJC), and tender joint counts (TJC) compared with placebo. Specifically, a significant improvement in DAS28-CRP was observed with combined namilumab doses versus placebo at Day 43 (p=0.0117) and Day 99 (p=0.0154).[11]

Building on these findings, a Phase 2 trial (NCT02393378, also known as NEXUS or EUCTR2013-002805-76-GB) was conducted. This 24-week, placebo-controlled study assessed three SC doses of namilumab (20 mg, 80 mg, and 150 mg) administered at baseline, week 2, week 6, and week 10, in combination with methotrexate, in 108 subjects with moderate to severe RA who had an inadequate response to methotrexate (MTX-IR) or a TNF inhibitor (TNF-IR).[14] The primary endpoint was the mean change from baseline in DAS28-CRP at week 12. The 150 mg dose of namilumab demonstrated a statistically significant difference in DAS28-CRP change compared to placebo at week 12 (p=0.005), with separation from placebo observed as early as week 2. Patients receiving 150 mg namilumab also showed higher ACR50 response rates at week 12.[14]

Despite these positive Phase 1b and Phase 2 results, Takeda, a key developer at the time, did not list namilumab in its subsequent pipeline updates for RA, suggesting a de-prioritization.[6] Ultimately, development for RA was discontinued.[4] The precise reasons for this discontinuation by Takeda or subsequent developers are not fully detailed in the provided information, but it often relates to a combination of factors including the overall risk-benefit profile in the context of existing therapies, strategic portfolio decisions, or challenges in demonstrating a sufficiently differentiated efficacy in larger, longer-term studies.

B. Plaque Psoriasis

Namilumab was investigated for moderate-to-severe plaque psoriasis in the Phase 2 NEPTUNE study (NCT02129777). This multicentre, randomized, double-blind, placebo-controlled, parallel-group, dose-finding trial evaluated four SC doses of namilumab (20 mg, 50 mg, 80 mg, and 150 mg) against placebo in 122 patients.[9] The primary endpoint was the proportion of patients achieving at least a 75% reduction in the Psoriasis Area and Severity Index (PASI 75) at week 12.[19]

The NEPTUNE study failed to meet its primary endpoint. The PASI 75 response rate at week 12 was low across all treatment groups, and no statistically significant difference was observed between any namilumab dose and placebo. Similar negative outcomes were noted for other clinical endpoints. Furthermore, laboratory investigations revealed no significant treatment-related changes from baseline in cell types, subpopulations, or cytokines relevant to the inflammatory pathways in psoriasis.[13]

The lack of efficacy was attributed to the understanding that GM-CSF blockade is likely not critical for suppressing the key inflammatory pathways driving psoriasis. The cytokine milieu in psoriasis, predominantly driven by IL-17A and IL-23, differs significantly from that in RA, where GM-CSF might play a more central role. It was considered unlikely that higher doses of namilumab would have led to a different outcome, given the flat dose-response observed.[13] Consequently, the development of namilumab for plaque psoriasis was discontinued.[4]

C. Axial Spondyloarthritis (Ankylosing Spondylitis - AS)

Izana Bioscience pursued the development of namilumab for axial spondyloarthritis, a severe form of arthritis affecting the spine. The Phase 2a NAMASTE study (NCT03622658) was a randomized, double-blind, placebo-controlled, proof-of-concept Bayesian trial conducted in the UK.[6] It enrolled 42 adult patients with moderate-to-severe active AS (meeting ASAS classification criteria, MRI criteria, and having an active Bath Ankylosing Spondylitis Disease Activity Index) who had an inadequate response or intolerance to at least one anti-TNF agent. Participants received SC namilumab 150 mg or placebo at weeks 0, 2, 6, and 10, with a 6:1 randomization ratio favoring namilumab.[15]

The primary endpoint was the proportion of participants achieving an ASAS20 (Assessment in SpondyloArthritis international Society 20% improvement) clinical response at week 12.[6] The NAMASTE trial did not meet its primary endpoint. At week 12, the ASAS20 response rate was numerically lower in the namilumab group (14 out of 36 patients; 38.9%) compared to the placebo group (3 out of 6 patients; 50%). The Bayesian posterior probability (η) was 0.72, which did not meet the pre-specified threshold of >0.927 for suggesting high clinical significance.[15] Given these results, further development for AS was not pursued. The small placebo group size (n=6), a feature of some Bayesian trial designs, can make interpretation challenging, particularly when the active arm underperforms relative to placebo.

D. Sarcoidosis (Pulmonary and Cardiac)

Kinevant Sciences, after acquiring Izana, investigated namilumab for sarcoidosis, a rare inflammatory disease characterized by granuloma formation. The Phase 2 RESOLVE-Lung study (NCT05314517) evaluated once-monthly SC namilumab in participants with chronic active pulmonary sarcoidosis.[9] The primary endpoint was the proportion of subjects experiencing a "Rescue Event" (defined as clinically significant worsening of sarcoidosis requiring treatment, failure to adhere to a protocol-defined oral corticosteroid taper, or premature discontinuation due to lack of benefit) during the double-blind period.[12]

In December 2024, it was announced that the RESOLVE-Lung study failed to meet its primary endpoint. Namilumab did not demonstrate a statistically significant treatment benefit compared to placebo. Secondary efficacy endpoints, including change in percent predicted forced vital capacity (%p FVC), corticosteroid tapering success, and change in the patient-reported King's Sarcoidosis Questionnaire, also failed to show a treatment benefit consistent with the primary endpoint.[12] Consequently, Kinevant Sciences discontinued the further development of namilumab for the treatment of sarcoidosis.[4]

A separate Phase 2 study, RESOLVE-Heart (NCT05351554), was initiated to evaluate namilumab in cardiac sarcoidosis.[9] However, this trial was terminated in 2022 due to a "sponsor business decision not related to safety".[7]

E. COVID-19 Pneumonia

During the COVID-19 pandemic, namilumab was investigated as a potential immunomodulator for severe COVID-19 pneumonia. The CATALYST trial, a Phase 2 randomized, open-label, multi-arm, adaptive, proof-of-concept study, was conducted in hospitalized patients (ward or ICU) in the UK with COVID-19 pneumonia and a C-reactive protein (CRP) level > 40mg/L.[5] Participants received either usual care (including steroids and oxygen/ventilation) or usual care plus a single intravenous (IV) dose of 150 mg namilumab. The primary endpoint focused on the improvement in inflammation, specifically the reduction in CRP concentration over time.[27]

The results showed that namilumab provided proof-of-concept evidence for reducing inflammation. There was a 97% probability of CRP being reduced over time in patients who received namilumab compared with those receiving usual care alone. Additionally, by day 28, there were fewer deaths (11% in the namilumab group vs. 19% in the usual care group) and more discharges from the hospital or ICU (78% vs. 61%, respectively) in the namilumab arm.[5] While these clinical outcome trends were positive, the study's sample size was too small for a definitive assessment of clinical efficacy. The investigators concluded that namilumab should be prioritized for further investigation in COVID-19.[5] However, as of December 2024, AdisInsight reported "No development reported" for namilumab in COVID-19 infections, suggesting that large-scale Phase 3 trials were not pursued or publicly announced.[4]

The pattern of Namilumab's clinical trial outcomes—consistent failure in chronic inflammatory diseases despite some early positive signals in RA, contrasted with promising signals in acute COVID-19 pneumonia—suggests that the therapeutic impact of GM-CSF neutralization may be highly context-dependent. GM-CSF's role might be more critical and therapeutically accessible in acute, intense inflammatory responses characterized by rapid CRP elevation, rather than in the complex, established immunopathology of chronic diseases where multiple redundant pathways and structural changes may be at play. Furthermore, the challenge of translating biomarker efficacy (like CRP reduction in COVID-19) into robust, statistically significant clinical benefit in larger, more heterogeneous populations remains a significant hurdle in drug development, as underscored by the discontinuation for sarcoidosis despite enrolling patients with confirmed active disease.[12]

IV. Pharmacokinetics (PK)

A. Summary of Available Pharmacokinetic Data

The pharmacokinetic profile of namilumab has been characterized in early-phase clinical trials. The Phase 1b PRIORA study in RA patients described its PK as linear and typical of an IgG1 monoclonal antibody following subcutaneous administration.[11]

More detailed PK data comes from a Phase 1 study (NCT02354599) conducted in healthy Japanese and Caucasian men aged 20-45 years.[30] In this study, Japanese subjects received single SC doses of namilumab at 80 mg, 150 mg, or 300 mg, while Caucasian subjects received 150 mg. The results indicated that the area under the serum concentration-time curve from zero to infinity (AUC0−∞​) and the maximum serum concentration (Cmax​) increased in a dose-proportional manner in Japanese subjects. For the 150 mg dose, the AUC0−∞​ was similar between Japanese subjects (575.2 µg·day/mL) and Caucasian subjects (559.7 µg·day/mL). However, the Cmax​ was approximately 40% higher in Japanese subjects receiving the 150 mg dose compared to their Caucasian counterparts. Despite this difference in Cmax​, the mean plasma total GM-CSF concentration-time profiles (a pharmacodynamic marker) were similar in both Japanese and Caucasian groups receiving 150 mg. This suggests that the observed Cmax​ difference did not translate into significant disparities in overall drug exposure or target engagement at this dose level and was not associated with notable safety concerns.[30] Specific values for Tmax​ (time to maximum concentration) and half-life (t1/2​) are not consistently detailed across the provided study summaries.[14] The overall PK profile appeared generally consistent and comparable between these ethnic groups, implying that PK variability was unlikely a primary cause for the mixed efficacy results seen in later, broader clinical trials.

B. Immunogenicity

The potential for immunogenicity, i.e., the development of anti-drug antibodies (ADAs), is an important consideration for all therapeutic proteins. For namilumab, early studies have reported low immunogenicity. In the Phase 1b PRIORA study in RA patients, no anti-namilumab antibodies were detected.[11] Similarly, in the Phase 1 study involving healthy Japanese and Caucasian men, namilumab induced no clinically relevant antibody response.[31] This consistent lack of significant ADA development in early trials is a favorable characteristic for a human monoclonal antibody, as ADAs can potentially neutralize drug activity, alter pharmacokinetics, lead to a loss of efficacy, or cause adverse events. The low immunogenicity observed suggests that the efficacy challenges encountered by namilumab in chronic disease trials were more likely due to the drug not adequately modulating the core disease pathology rather than being compromised by an immune response against the antibody itself.

V. Safety and Tolerability Profile

A. Overview of Common and Serious Adverse Events (AEs)

Namilumab has generally been reported as well tolerated across its clinical development program, including studies in rheumatoid arthritis, plaque psoriasis, axial spondyloarthritis, sarcoidosis, healthy volunteers, and COVID-19 pneumonia.[6]

In the Phase 1b PRIORA study for mild-to-moderate RA, the incidence of treatment-emergent adverse events (TEAEs) was similar between namilumab-treated groups (150 mg: 63%; 300 mg: 57%) and the placebo group (56%). The most frequently reported TEAEs (≥10% overall) were nasopharyngitis (17%) and exacerbation or worsening of RA (13%).[11]

In the Phase 2 study (NCT02393378) for moderate-to-severe RA, the most common TEAEs (across placebo, 20 mg, 80 mg, and 150 mg namilumab arms, respectively) included nasopharyngitis (18.5%, 17.9%, 4.0%, 14.3%), dyspnoea (0.0%, 3.6%, 8.0%, 10.7%), bronchitis (7.4%, 3.6%, 4.0%, 3.6%), and headache (3.7%, 3.6%, 12.0%, 0.0%). One serious adverse event, a myocardial infarction, was observed in the 150 mg namilumab group. No serious infections were reported, and there was no apparent dose relationship for AEs.[14]

In healthy Japanese and Caucasian volunteers (NCT02354599), AEs were generally mild to moderate, with no dose-proportional increase noted in Japanese subjects.[30]

For the NAMASTE trial in axial spondyloarthritis, the rates of any TEAEs were reported as similar between the namilumab and placebo groups (31 events in 36 namilumab patients vs. 5 events in 6 placebo patients, noting the imbalance in group sizes).[15]

In the RESOLVE-Lung study for pulmonary sarcoidosis, the safety profile of namilumab was consistent with that observed in previous studies, with no new safety signals emerging that led to the trial's discontinuation; the discontinuation was due to lack of efficacy.[12]

In the CATALYST trial for COVID-19 pneumonia, 134 AEs occurred in 30 out of 55 patients (55%) in the namilumab group, compared to 145 AEs in 29 out of 54 patients (54%) in the usual care group. The mortality rate was 11% (6 out of 57 patients) in the namilumab group versus 19% (10 out of 54 patients) in the usual care group by day 28. Safety data did not suggest an increased risk of secondary infections with namilumab.[27]

This consistent safety profile across diverse patient populations and conditions suggests that the repeated discontinuation of Namilumab development programs was primarily driven by insufficient efficacy rather than by prohibitive safety concerns.

B. Known or Potential Drug Interactions

Information from DrugBank indicates potential pharmacodynamic interactions for namilumab.[1] The risk or severity of adverse effects may be increased when namilumab is combined with a wide array of other monoclonal antibodies. This is a general consideration for combination biologic therapies, potentially due to cumulative immunosuppressive effects or broader modulation of the immune system. Examples of such antibodies listed include Amivantamab, Anifrolumab, Avelumab, Basiliximab, Bimekizumab, Canakinumab, Daratumumab, and Dinutuximab, among many others.[1]

Additionally, certain estrogenic compounds, such as Diethylstilbestrol, Esterified estrogens, Estetrol, Estradiol (and its various esters like acetate, benzoate, cypionate, valerate), and Estriol, are noted to potentially increase the thrombogenic activities of Namilumab.[1] This is a class consideration for some biologics when co-administered with hormonal agents. However, the clinical trial summaries provided for namilumab did not prominently feature adverse events specifically attributed to these drug-drug interactions as major factors leading to study discontinuations, suggesting they were either managed effectively, infrequently encountered, or not severe enough to halt development independently.

VI. Development History and Regulatory Status

A. Key Developers and Licensing Agreements

Namilumab's development journey has involved several pharmaceutical companies through licensing agreements and acquisitions. The drug was originated by Micromet Inc..[4] In 2012, Amgen acquired Micromet.[6] Subsequently, Takeda Pharmaceuticals International obtained rights to namilumab and conducted clinical trials, notably in rheumatoid arthritis and plaque psoriasis.[4]

Around 2017, Izana Bioscience licensed the exclusive worldwide rights from Takeda to develop, manufacture, and commercialize namilumab (then referred to as IZN-101 by Izana) across all indications. Takeda retained a strategic equity stake in Izana. Izana's initial focus was on developing namilumab for ankylosing spondylitis, supported by a £1.35 million grant from Innovate UK, the UK's innovation agency.[4]

Later, Kinevant Sciences, a biopharmaceutical company under the Roivant Sciences umbrella, acquired Izana Bioscience and continued the development of namilumab, primarily focusing on sarcoidosis.[4]

Table 3: Overview of Namilumab Developers and Licensing

CompanyRoleKey Development Period / Indications Focused OnSignificant Licensing Events/NotesSnippet References
Micromet Inc.OriginatorEarly Development4
AmgenAcquirer of OriginatorPost-acquisition phaseAcquired Micromet in 20126
Takeda Pharmaceuticals InternationalDeveloper/Licensee~2011-2017 / Rheumatoid Arthritis, Plaque PsoriasisConducted Phase 1b/2 trials4
Izana BioscienceDeveloper/Licensee~2017- acquisition by Kinevant / Ankylosing SpondylitisLicensed worldwide rights from Takeda; Received Innovate UK grant4
Kinevant Sciences (Roivant)Developer/LicenseePost-acquisition of Izana - 2024 / Sarcoidosis (Pulmonary, Cardiac)Acquired Izana Bioscience4

B. Chronology of Development and Discontinuation Across Indications

Namilumab's clinical development commenced with Takeda investigating its potential in rheumatoid arthritis (Phase 1b PRIORA, NCT01317797, initiated around 2011; Phase 2 NEXUS, NCT02393378, initiated around 2014-2015) and plaque psoriasis (Phase 2 NEPTUNE, NCT02129777, initiated around 2014).[9]

The development for plaque psoriasis was halted following the failure of the NEPTUNE trial to demonstrate efficacy (results published around 2019).[4] Similarly, development for rheumatoid arthritis was also discontinued by Takeda and subsequent developers.[4]

Izana Bioscience licensed namilumab around 2017-2018 and initiated the NAMASTE trial (NCT03622658) in 2018 for axial spondyloarthritis. This trial also failed to meet its efficacy endpoints, with results becoming available around 2024.[6]

Kinevant Sciences then took over development, focusing on sarcoidosis. The RESOLVE-Lung trial (NCT05314517) for pulmonary sarcoidosis was initiated around 2022 but failed to meet its endpoints, leading to the discontinuation of development for sarcoidosis in December 2024.[4] The RESOLVE-Heart trial (NCT05351554) for cardiac sarcoidosis, also initiated around 2022, was terminated early in the same year due to a sponsor business decision unrelated to safety.[7]

During the pandemic (around 2020-2021), namilumab was investigated in the CATALYST trial for COVID-19 pneumonia, where it showed some promising anti-inflammatory effects and was recommended for further study.[5] However, its current development status for COVID-19 is listed as "no development reported" by AdisInsight as of December 2024.[4]

This multi-company development path, characterized by licensing and acquisitions, is indicative of assets that possess initial biological plausibility but face significant challenges in demonstrating definitive clinical efficacy. Each successive company likely aimed to identify a suitable indication or an optimized trial design, yet the fundamental efficacy limitations in chronic conditions appear to have persisted. The Innovate UK grant for Izana's AS trial underscores how external funding can support development in areas of unmet need, but ultimate success hinges on the scientific and clinical validation of the therapeutic approach.[21] The eventual discontinuation for Sarcoidosis by Kinevant, a company specializing in rare inflammatory diseases, further reinforces the significant hurdles Namilumab faced in this therapeutic area.[23]

C. Current Overall Status of Namilumab

As of late 2024, namilumab has been largely discontinued for the chronic inflammatory diseases for which it was extensively studied, including plaque psoriasis, rheumatoid arthritis, axial spondyloarthritis, and sarcoidosis, primarily due to insufficient efficacy in Phase 2 trials.[4] While early signals in COVID-19 pneumonia were positive, leading to recommendations for further investigation, there is no current information in the provided materials to suggest that Phase 3 development for COVID-19 was initiated or is ongoing.[4]

VII. Expert Analysis and Conclusion

A. Synthesis of Namilumab's Development Journey

Namilumab, a human monoclonal antibody targeting GM-CSF, embarked on its development journey with a strong biological rationale for treating a spectrum of inflammatory and autoimmune diseases. Its trajectory—from initial promise grounded in its mechanism of action and early-phase data to subsequent difficulties in demonstrating robust, consistent efficacy in larger, controlled trials for complex chronic conditions—is a common narrative in pharmaceutical development. The molecule itself appears well-characterized with a generally manageable safety and pharmacokinetic profile.

B. Insights into Targeting GM-CSF

The clinical development of namilumab provides valuable insights into the therapeutic utility of targeting GM-CSF. Its failure to achieve primary endpoints in plaque psoriasis, axial spondyloarthritis, and sarcoidosis suggests that while GM-CSF is undeniably a pro-inflammatory cytokine, its role may not be universally pivotal, or its neutralization alone may be insufficient to achieve clinical remission in all chronic inflammatory states.[13] The distinct pathophysiological mechanisms and cytokine profiles driving different diseases, as highlighted by the differing roles of IL-17/IL-23 in psoriasis versus potentially IL-6 in RA, are critical factors.[13] This underscores that a "one-target-fits-all-inflammatory-diseases" strategy is rarely successful.

In contrast, the positive signals observed in the CATALYST trial for acute COVID-19 pneumonia, particularly the reduction in the inflammatory biomarker CRP and trends towards improved clinical outcomes, suggest that GM-CSF might play a more dominant and therapeutically accessible role in acute, severe hyperinflammatory responses.[5] This differential outcome emphasizes that the therapeutic window and impact of GM-CSF inhibition are highly dependent on the specific pathological state, disease duration, and the interplay of other immune mediators.

C. Evaluation of Existing Data and Knowledge Gaps

The available data consistently point to a favorable safety and pharmacokinetic profile for namilumab, which were generally not the limiting factors in its development. The primary knowledge gap revolves around the precise reasons for the lack of efficacy in chronic conditions despite its plausible mechanism. Potential contributing factors could include insufficient target engagement within specific tissue microenvironments, the presence of redundant or compensatory inflammatory pathways, inappropriate patient selection criteria that did not enrich for populations most likely to respond to GM-CSF blockade, or clinical endpoints that were not sensitive enough to detect more subtle, albeit potentially meaningful, effects.

A significant gap is the absence of publicly available Phase 3 data for any indication. Even for COVID-19, where Phase 2 results were encouraging, the progression to, and outcome of, larger confirmatory trials remains unreported in the provided materials.

D. Concluding Remarks

Namilumab's development history serves as an illustrative case study in the complexities of translating a targeted biological therapy into broad clinical success. The direct neutralization of GM-CSF by namilumab did not yield effective treatments for the chronic inflammatory diseases for which it was most extensively evaluated. Nevertheless, these studies contribute to a deeper understanding of GM-CSF's multifaceted roles in human health and disease.

The promising, albeit preliminary, findings in acute COVID-19 pneumonia hinted at a potential niche for GM-CSF blockade in specific hyperinflammatory states. However, without further development and data, this remains speculative. Overall, the journey of namilumab highlights the significant challenges inherent in drug development for complex immune-mediated diseases, emphasizing the need for nuanced patient stratification, a profound understanding of disease-specific biology, and perhaps combination strategies to effectively tackle such conditions. The "target-rich, translation-poor" phenomenon is evident, where a scientifically valid target like GM-CSF does not automatically translate into widespread therapeutic success due to the intricate and often redundant nature of biological systems in chronic disease.

Works cited

  1. Namilumab: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed June 12, 2025, https://go.drugbank.com/drugs/DB13037
  2. namilumab | Ligand page - IUPHAR/BPS Guide to PHARMACOLOGY, accessed June 12, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=clinical&ligandId=10370
  3. Namilumab Monoclonal Antibody - Ontosight.ai, accessed June 12, 2025, https://ontosight.ai/glossary/term/namilumab-monoclonal-antibody--67a16e8a6c3593987a5722b4
  4. Namilumab - Amgen/Izana Bioscience/Takeda - AdisInsight - Springer, accessed June 12, 2025, https://adisinsight.springer.com/drugs/800030372
  5. pubmed.ncbi.nlm.nih.gov, accessed June 12, 2025, https://pubmed.ncbi.nlm.nih.gov/34922649/#:~:text=Interpretation%3A%20Namilumab%2C%20but%20not%20infliximab,Funding%3A%20Medical%20Research%20Council.
  6. Izana starts proof-of-concept trial for spinal arthritis drug - Fierce Biotech, accessed June 12, 2025, https://www.fiercebiotech.com/biotech/izana-starts-proof-concept-trial-for-spinal-arthritis-drug
  7. Roivant subsidiary bins failed pulmonary programme - FirstWord Pharma, accessed June 12, 2025, https://firstwordpharma.com/story/5933930
  8. NAMILUMAB - GSRS, accessed June 12, 2025, https://gsrs.ncats.nih.gov/ginas/app/beta/substances/MED485W763
  9. Namilumab (AMG-203) | Anti-Human CSF2 Antibody | MedChemExpress, accessed June 12, 2025, https://www.medchemexpress.com/namilumab.html
  10. Namilumab Overview - Creative Biolabs, accessed June 12, 2025, https://www.creativebiolabs.net/namilumab-overview.htm
  11. (PDF) Phase 1b randomized, double-blind study of namilumab, an anti-granulocyte macrophage colony-stimulating factor monoclonal antibody, in mild-to-moderate rheumatoid arthritis - ResearchGate, accessed June 12, 2025, https://www.researchgate.net/publication/314718298_Phase_1b_randomized_double-blind_study_of_namilumab_an_anti-granulocyte_macrophage_colony-stimulating_factor_monoclonal_antibody_in_mild-to-moderate_rheumatoid_arthritis
  12. Roivant Announces Topline Results from Phase 2 RESOLVE-Lung Study of Namilumab in Chronic Active Pulmonary Sarcoidosis - BioSpace, accessed June 12, 2025, https://www.biospace.com/press-releases/roivant-announces-topline-results-from-phase-2-resolve-lung-study-of-namilumab-in-chronic-active-pulmonary-sarcoidosis
  13. Targeting granulocyte‐monocyte colony‐stimulating factor in psoriasis. What a negative study can teach us | British Journal of Dermatology | Oxford Academic, accessed June 12, 2025, https://academic.oup.com/bjd/article/180/6/1286/6731144
  14. Efficacy and safety of namilumab, a human monoclonal antibody ..., accessed June 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6471864/
  15. Granulocyte-macrophage colony-stimulating factor neutralisation in ..., accessed June 12, 2025, https://pubmed.ncbi.nlm.nih.gov/38942047/
  16. Phase 1b randomized, double-blind study of namilumab, an anti ..., accessed June 12, 2025, https://pubmed.ncbi.nlm.nih.gov/28274253/
  17. SAT0210 First-in-Patient Study of Namilumab, an Anti-GM-CSF Monoclonal Antibody, in Active Rheumatoid Arthritis: Results of the Priora Phase IB Study - ResearchGate, accessed June 12, 2025, https://www.researchgate.net/publication/281201687_SAT0210_First-in-Patient_Study_of_Namilumab_an_Anti-GM-CSF_Monoclonal_Antibody_in_Active_Rheumatoid_Arthritis_Results_of_the_Priora_Phase_IB_Study
  18. A 24-week placebo-controlled clinical study to evaluate the adequate dose, efficacy and safety of 3 doses of namilumab combined with methotrexate in subjects with moderate to severe rheumatoid arthritis. - MedPath Clinical Trial Search, accessed June 12, 2025, https://trial.medpath.com/clinical-trial/3f0a1761d92204e6
  19. Efficacy and Safety of Namilumab (MT203) for Plaque Psoriasis - ClinConnect, accessed June 12, 2025, https://clinconnect.io/trials/NCT02129777
  20. Granulocyte-macrophage colony-stimulating factor (GM-CSF) as a ..., accessed June 12, 2025, https://pubmed.ncbi.nlm.nih.gov/30207587/
  21. Izana Bioscience Initiates Namilumab Phase II Proof-of-Concept Clinical Study in Ankylosing Spondylitis - PR Newswire, accessed June 12, 2025, https://www.prnewswire.com/news-releases/izana-bioscience-initiates-namilumab-phase-ii-proof-of-concept-clinical-study-in-ankylosing-spondylitis-846730253.html
  22. Namilumab Fails to Meet Primary Endpoint in Phase 2 RESOLVE-Lung Study for Pulmonary Sarcoidosis - MedPath, accessed June 12, 2025, https://trial.medpath.com/news/0e9108c20f1ee904/kinevant-sciences-announces-topline-results-from-phase-2-resolve-lung-study-of-biospace
  23. Kinevant Sciences Announces Topline Results from Phase 2 RESOLVE-Lung Study of Namilumab in Chronic Active Pulmonary Sarcoidosis - BioSpace, accessed June 12, 2025, https://www.biospace.com/press-releases/kinevant-sciences-announces-topline-results-from-phase-2-resolve-lung-study-of-namilumab-in-chronic-active-pulmonary-sarcoidosis
  24. 0001140361-24-048238 | 8-K | iXBRL Viewer - Investor Relations | Roivant Sciences Ltd., accessed June 12, 2025, https://investor.roivant.com/node/11396/ixbrl-viewer
  25. Important Clinical Trial Results Announcement for Sarcoidosis Patients, accessed June 12, 2025, https://www.stopsarcoidosis.org/important-clinical-trial-results-announcement-for-sarcoidosis-patients/
  26. Namilumab Discontinued After Phase II Trial Failure for Pulmonary Sarcoidosis - Xtalks, accessed June 12, 2025, https://xtalks.com/namilumab-discontinued-after-phase-ii-trial-failure-for-pulmonary-sarcoidosis-3996/
  27. Namilumab or infliximab compared with standard of care in hospitalised patients with COVID-19 (CATALYST): a randomised, multicentre, multi-arm, multistage, open-label, adaptive, phase 2, proof-of-concept trial | Sciety, accessed June 12, 2025, https://sciety.org/articles/activity/10.1101/2021.06.02.21258204?utm_source=sciety_labs_article_page
  28. Izana Bioscience Namilumab May Treat COVID-19 Pneumonia ..., accessed June 12, 2025, https://respiratory-therapy.com/disorders-diseases/infectious-diseases/izana-biosciences-namilumab-covid-19-pneumonia/
  29. Namilumab or infliximab compared with standard of care in hospitalised patients with COVID-19 (CATALYST): a randomised, multicentre, multi-arm, multistage, open-label, adaptive, phase 2, proof-of-concept trial. - The Kennedy Institute of Rheumatology - University of Oxford, accessed June 12, 2025, https://www.kennedy.ox.ac.uk/@@disable-cookies?came_from=https://www.kennedy.ox.ac.uk/publications/1225685
  30. Namilumab - Semantic Scholar, accessed June 12, 2025, https://www.semanticscholar.org/topic/Namilumab/2171881
  31. Randomized, double-blind, placebo-controlled, phase I study of the safety and pharmacokinetics of namilumab in healthy Japanese and Caucasian men - PubMed, accessed June 12, 2025, https://pubmed.ncbi.nlm.nih.gov/30168415/
  32. Izana Bioscience Launches as Clinical-Stage Translational Medicine Company, accessed June 12, 2025, https://www.pharmoutsourcing.com/1315-News/345357-Izana-Bioscience-Launches-as-Clinical-Stage-Translational-Medicine-Company/?catid=6266
  33. Namilumab (AMG-203; Anti-Human CSF2 Recombinant Antibody) | CAS 1206681-39-1, accessed June 12, 2025, https://www.abmole.com/products/namilumab.html

Published at: June 12, 2025

This report is continuously updated as new research emerges.

© Copyright 2025. All Rights Reserved by MedPath