Lirentelimab, also known as AK002, is an investigational biotech drug developed by Allakos Inc. as a humanized, afucosylated IgG1 monoclonal antibody designed to target Sialic acid-binding immunoglobulin-like lectin 8 (Siglec-8).[1] The therapeutic rationale for Lirentelimab was based on a novel and elegant dual mechanism of action targeting key effector cells in Type 2 inflammation. By binding to Siglec-8, a receptor selectively expressed on eosinophils and mast cells, Lirentelimab was engineered to deplete pathogenic eosinophils through antibody-dependent cell-mediated cytotoxicity (ADCC) and simultaneously inhibit the activation and degranulation of mast cells.[1]
This dual-pronged approach held significant promise for a wide range of allergic and inflammatory conditions where these cells are implicated, including eosinophilic gastrointestinal diseases (EGIDs), atopic dermatitis (AD), and chronic spontaneous urticaria (CSU). Early and mid-stage clinical trials showed that Lirentelimab was exceptionally effective at its biological function, consistently demonstrating profound and sustained depletion of eosinophils in both blood and tissue across multiple patient populations.[7]
However, the clinical development program was ultimately defined by a critical and recurring paradox: the drug's potent histologic and pharmacodynamic activity repeatedly failed to translate into statistically significant improvements in patient-reported symptoms. While an initial Phase 2 study in eosinophilic gastritis and duodenitis (ENIGMA) showed promising alignment between eosinophil reduction and symptom improvement, subsequent larger, pivotal trials (ENIGMA 2 and KRYPTOS) in EGIDs failed to meet their symptomatic co-primary endpoints despite achieving overwhelming statistical success on their histologic co-primary endpoints.[7]
A subsequent strategic pivot to dermatologic indications proved equally unsuccessful. Phase 2 trials in atopic dermatitis (ATLAS) and chronic spontaneous urticaria (MAVERICK) also failed to demonstrate a clinical benefit over placebo, despite again confirming near-complete depletion of peripheral eosinophils.[8] These definitive failures led Allakos Inc. to announce the complete discontinuation of the Lirentelimab development program in January 2024.[11] The journey of Lirentelimab serves as a critical case study in drug development, highlighting the complex and often unpredictable relationship between surrogate biomarkers, such as eosinophil counts, and clinically meaningful outcomes, thereby challenging foundational assumptions about the pathophysiology of several major inflammatory diseases.
The development of Lirentelimab represents a targeted therapeutic strategy rooted in a sophisticated understanding of the cellular drivers of allergic and inflammatory diseases. Its creation was predicated on the identification of a unique cell surface receptor, Siglec-8, that offered a precise way to modulate the two primary effector cells of Type 2 immunity: eosinophils and mast cells.
Eosinophils and mast cells are key cellular components of the immune system, centrally involved in the pathogenesis of a wide spectrum of inflammatory conditions, often categorized under the umbrella of Type 2 or T-helper 2 (Th2) cell-mediated immunity.[13] These diseases include eosinophilic gastrointestinal diseases (EGIDs) such as eosinophilic esophagitis (EoE), eosinophilic gastritis (EG), and eosinophilic duodenitis (EoD), as well as atopic dermatitis (eczema), asthma, and chronic urticaria.[1]
In these conditions, inappropriate activation and accumulation of eosinophils and mast cells in specific tissues drive chronic inflammation and subsequent organ damage. Eosinophils, when activated, release a payload of cytotoxic granule proteins, reactive oxygen species, and pro-inflammatory cytokines that can cause direct tissue injury. Mast cells, upon activation (often via IgE-dependent pathways), rapidly degranulate, releasing potent mediators like histamine, prostaglandins, and leukotrienes, which cause vasodilation, smooth muscle contraction, and sensory nerve activation.[1] The clinical manifestations of these cellular processes are the debilitating symptoms experienced by patients, including severe abdominal pain and cramping in EGIDs, dysphagia (difficulty swallowing) in EoE, and intense pruritus (itching) and wheals in atopic dermatitis and urticaria.[14]
Traditional treatments for these conditions have often relied on broad-spectrum immunosuppressants like corticosteroids, which, while effective, are associated with significant long-term side effects.[14] The advent of biologics marked a shift towards more targeted therapies, such as those blocking key cytokines like Interleukin-5 (IL-5), which is critical for eosinophil survival and differentiation. The development of Lirentelimab represented a further evolution of this targeted approach, moving beyond the modulation of signaling pathways to the direct engagement and manipulation of the pathogenic cells themselves.
Sialic acid-binding immunoglobulin-like lectin 8 (Siglec-8) is a member of the Siglec family of inhibitory I-type lectins.[3] Its therapeutic appeal stems from its highly restricted expression profile. Siglec-8 is found almost exclusively on the surface of human eosinophils and mast cells, with only low-level expression reported on basophils.[1] This cellular specificity makes it an ideal target for therapeutic intervention, as it allows for the precise modulation of the key pathogenic cells while sparing other components of the immune system, thereby minimizing the potential for off-target effects.[3]
Functionally, Siglec-8 is an inhibitory receptor. Its intracellular domain contains immunoreceptor tyrosine-based inhibitory motifs (ITIMs). When Siglec-8 is engaged by a ligand or an agonistic antibody, these ITIMs become phosphorylated and recruit phosphatases, such as SHP-1. These phosphatases counteract the activating signals driven by kinase signaling cascades within the cell, effectively acting as a "brake" on cellular activation.[3] This natural immunomodulatory function provided a clear rationale for developing a therapeutic agent that could artificially engage this receptor to control eosinophil and mast cell activity.
Lirentelimab (AK002) was engineered as a humanized IgG1 monoclonal antibody designed to bind specifically to Siglec-8 and function as an agonist, triggering its inhibitory signaling pathways and other cellular responses.[1] A key structural feature of Lirentelimab is that it is afucosylated. This means it lacks fucose sugar residues in the Fc (constant) region of the antibody. This specific glycoengineering modification significantly enhances the antibody's binding affinity for the Fc-gamma receptor IIIa (), which is expressed on the surface of Natural Killer (NK) cells.[3] The enhanced binding potentiates the ability of Lirentelimab to induce a powerful cytotoxic immune response known as antibody-dependent cell-mediated cytotoxicity (ADCC).[5]
The central therapeutic hypothesis for Lirentelimab was therefore a powerful, dual-pronged mechanism of action. By binding to Siglec-8 on eosinophils, the afucosylated Fc region would recruit NK cells to induce ADCC, leading to the rapid and efficient depletion of these tissue-damaging cells. Simultaneously, by binding to Siglec-8 on mast cells, the antibody would trigger the receptor's intrinsic inhibitory function, preventing their degranulation and the release of inflammatory mediators.[3] This combined approach of eliminating one pathogenic cell type while pacifying another was envisioned as a highly effective strategy for treating a broad array of eosinophil- and mast cell-mediated diseases.
| Property | Value | Source(s) |
|---|---|---|
| Generic Name | Lirentelimab | 18 |
| Development Name | AK002 | 1 |
| DrugBank ID | DB16728 | User Query |
| CAS Number | 2283348-97-8 | 1 |
| Type | Biotech, Humanized Monoclonal Antibody | 1 |
| Isotype | Human IgG1, kappa | 20 |
| Molecular Formula | 1 | |
| Molecular Weight | ~144.3 kDa - 150 kDa | 1 |
| Target | SIGLEC8 (Sialic acid-binding Ig-like lectin 8) | 1 |
The foundation of Lirentelimab's clinical development program was built upon its well-defined molecular characteristics and a robust body of preclinical evidence that validated its proposed dual mechanism of action.
Lirentelimab is a large-molecule biologic drug, classified as a humanized IgG1 kappa monoclonal antibody.[1] Its fundamental identifiers include the DrugBank ID DB16728, CAS Registry Number 2283348-97-8, and United States Adopted Name (USAN) File Number GH-173.[23] The molecular structure is complex, with a chemical formula of and a corresponding molecular weight of approximately 144.3 kDa to 150 kDa.[1] As a peptide-based therapeutic, its structure is defined by the specific amino acid sequence of its heavy and light chains.[18] For clinical and commercial production, Lirentelimab was manufactured using recombinant DNA technology in Chinese Hamster Ovary (CHO) cells, a standard bioprocessing platform for monoclonal antibodies.[20]
The therapeutic potential of Lirentelimab was derived from its ability to exert two distinct and complementary effects on the primary cellular drivers of allergic inflammation.
Lirentelimab was designed to be a potent eosinophil-depleting agent. Upon binding to Siglec-8 on the surface of an eosinophil, it initiates cell death through two synergistic pathways.
The primary mechanism is ADCC. The afucosylated Fc region of the Lirentelimab molecule binds with high affinity to receptors on NK cells. This cross-linking acts as a bridge, bringing the NK cell into close proximity with the eosinophil and activating the NK cell to release cytotoxic proteins like perforin and granzymes, which induce lysis and death of the targeted eosinophil.[1] This process is highly efficient and is the principal driver of rapid eosinophil clearance from the circulation and tissues.
In addition to ADCC, Lirentelimab can induce direct programmed cell death, or apoptosis, in eosinophils.[4] In vitro studies demonstrated that cross-linking of Siglec-8 receptors on the eosinophil surface initiates an apoptotic cascade.[15] This effect is significantly amplified when the eosinophils are in a "primed" state, which occurs in the presence of pro-inflammatory cytokines such as IL-5, IL-33, and granulocyte-macrophage colony-stimulating factor (GM-CSF).[1] Since these cytokines are abundant in the microenvironment of inflamed tissues characteristic of diseases like EGIDs and asthma, this suggests that Lirentelimab's apoptotic activity would be most potent precisely where it is needed most. In this cytokine-primed state, the cell death mechanism shifts to a caspase-independent pathway involving mitochondrial damage and the production of reactive oxygen species (ROS) via the nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase) enzyme complex.[15]
In stark contrast to its effect on eosinophils, Lirentelimab's engagement of Siglec-8 on mast cells is inhibitory, not cytotoxic.[6] Binding of Lirentelimab to mast cell Siglec-8 triggers the receptor's intrinsic inhibitory function by activating the intracellular ITIM signaling cascade.[3] This intracellular signal actively suppresses mast cell activation, particularly in response to IgE-mediated stimuli. Consequently, Lirentelimab inhibits the degranulation of mast cells and prevents the release of a host of potent inflammatory mediators, including histamine and prostaglandin D2 ().[1] This mechanism does not reduce mast cell numbers through direct killing but rather pacifies them, reducing their contribution to the inflammatory milieu. Preclinical studies in humanized mouse models provided compelling in vivo validation of this effect, showing that administration of a Siglec-8 antibody could protect the animals from systemic anaphylaxis, a condition driven by widespread mast cell degranulation.[4]
This dual mechanism was the cornerstone of Lirentelimab's therapeutic proposition. It offered the potential to not only remove a key source of chronic tissue damage (eosinophils) but also to quell the acute, symptom-driving inflammatory bursts from mast cells. However, this elegant design also introduced a layer of complexity. The success of the therapy depended on the assumption that both eosinophil-driven damage and mast cell-driven symptoms were critical, and that both mechanisms would be equally effective and relevant in human disease. The subsequent clinical trial results would ultimately challenge these core assumptions.
The decision to advance Lirentelimab into human clinical trials was supported by a strong preclinical data package. In vitro experiments established its high binding affinity, with the monovalent fragment antigen-binding (Fab) portion of the antibody binding to the recombinant extracellular domain of Siglec-8 with an affinity of 464 pM.[4] The antibody demonstrated selective binding to eosinophils and mast cells in human peripheral blood and lung tissue samples.[4] Its functional activity was confirmed in cell-based assays, which showed potent ADCC activity against human eosinophils, with a half-maximal effective concentration () of just 1.9 ng/mL.[4] In vivo studies in animal models further substantiated the therapeutic concept, demonstrating both the depletion of eosinophils and the inhibition of mast cell-mediated anaphylactic responses.[4] These preclinical findings provided a robust rationale for investigating Lirentelimab as a novel treatment for eosinophil- and mast cell-mediated diseases in humans.
The clinical development of Lirentelimab in EGIDs represents a compelling narrative of initial triumph followed by a profound and unexpected setback. The journey through Phase 2 and Phase 3 trials in this indication was pivotal, ultimately revealing a critical disconnect between the drug's biological activity and its clinical efficacy that would define its legacy.
| Trial Name/Identifier (NCT) | Phase | Indication(s) | Status | Key Outcome Summary |
|---|---|---|---|---|
| ENIGMA (NCT03496571) | 2 | Eosinophilic Gastritis / Duodenitis (EG/EoD) | Completed | Positive: Met primary (histologic) and secondary (symptomatic) endpoints.24 |
| ENIGMA 2 (NCT04322604) | 3 | EG/EoD | Completed | Negative: Met histologic co-primary endpoint but missed symptomatic co-primary endpoint.7 |
| KRYPTOS (NCT04322708) | 2/3 | Eosinophilic Esophagitis (EoE) | Completed | Negative: Met histologic co-primary endpoint but missed symptomatic co-primary endpoint.7 |
| ATLAS (NCT05155085) | 2 | Atopic Dermatitis (AD) | Terminated | Negative: Failed to meet primary endpoint (EASI-75).8 |
| MAVERICK | 2b | Chronic Spontaneous Urticaria (CSU) | Terminated | Negative: Failed to meet primary endpoint (UAS7).8 |
| NCT02808793 | 1 | Indolent Systemic Mastocytosis (ISM) | Completed | Positive: Generally well-tolerated with significant improvement in symptoms and quality of life.31 |
| NCT03379311 | 1 | Allergic Conjunctivitis | Completed | Positive: Well-tolerated with preliminary efficacy signals.33 |
The clinical journey of Lirentelimab began with remarkable promise in the Phase 2 ENIGMA study (NCT03496571). This randomized, double-blind, placebo-controlled trial enrolled 65 adults with symptomatic EG and/or EoD.[25] The results, published in the prestigious New England Journal of Medicine, provided strong proof-of-concept for the drug's therapeutic hypothesis.[24]
The trial successfully met its primary endpoint, demonstrating a profound reduction in tissue eosinophil counts. Patients receiving Lirentelimab experienced a mean reduction of 86% in gastrointestinal eosinophils from baseline, compared to a 9% increase in the placebo group ().[25] Crucially, this potent histologic effect was accompanied by significant symptomatic improvement. The study also met its key secondary endpoints:
This alignment of robust histologic clearance with statistically significant and clinically meaningful symptom relief was the ideal outcome for a Phase 2 study. It strongly suggested that eosinophil depletion was directly linked to patient well-being and provided a clear and compelling rationale for advancing Lirentelimab into larger, registrational Phase 3 trials.[14]
The optimism generated by the ENIGMA study was decisively challenged by the results of the subsequent, larger pivotal trials: the ENIGMA 2 Phase 3 study in EG/EoD (NCT04322604) and the KRYPTOS Phase 2/3 study in EoE (NCT04322708).[2] These studies were designed with co-primary endpoints, a common requirement for regulatory approval in many chronic diseases, mandating success on both a biological marker (histology) and a patient-reported outcome (symptoms).[7] While Lirentelimab demonstrated overwhelming success on the first, it failed unequivocally on the second, revealing a stunning disconnect that would prove fatal to its development in EGIDs.
Across both large-scale trials, Lirentelimab replicated the profound eosinophil-depleting effect seen in Phase 2. The drug was exceptionally effective at clearing eosinophils from the inflamed gastrointestinal tissue, meeting the histologic co-primary endpoints with high statistical significance.
These results confirmed that Lirentelimab possessed a powerful and consistent biological effect on its intended target cell population in the diseased tissue.
Despite the dramatic histologic improvements, neither trial succeeded in demonstrating a corresponding benefit in patient-reported symptoms, thereby failing their symptomatic co-primary endpoints.
This stark contradiction between the Phase 2 ENIGMA results and the outcomes of the larger pivotal trials raised critical questions. The unexpectedly high placebo response on subjective symptom scores in the longer, larger trials may have obscured a modest drug effect. Alternatively, it suggested that the initial positive symptom signal in Phase 2 may have been an anomaly. Most importantly, the results severely challenged the foundational "eosinophil-as-driver" hypothesis for symptoms in EGIDs. The data strongly implied that simply removing eosinophils from the tissue, even to near-complete levels, was insufficient to alleviate the symptoms that matter most to patients. This finding has profound implications, not just for Lirentelimab, but for the entire field of drug development in EGIDs, forcing a re-evaluation of whether eosinophil counts are an appropriate surrogate endpoint for clinical benefit.
| Endpoint | Trial | Lirentelimab Arm(s) | Placebo Arm | P-value |
|---|---|---|---|---|
| Histologic Endpoint (% Responders) | ENIGMA 2 (EG/EoD) | 84.6% | 4.5% | |
| KRYPTOS (EoE) | 87.9% (High Dose) 92.5% (Low Dose) | 10.9% | ||
| Symptomatic Endpoint (Mean Change from Baseline) | ENIGMA 2 (TSS-6 Score) | -10.0 | -11.5 | |
| KRYPTOS (DSQ Score) | -17.4 (High Dose) -11.9 (Low Dose) | -14.6 | ||
| Data sourced from.7 | ||||
An open-label extension study (NCT04620811) was conducted to evaluate the long-term safety and efficacy for patients who completed the pivotal EG/EoD trials.[26] While this study provided valuable long-term safety data, the failure of the parent trials to meet their primary symptomatic endpoints rendered the extension data moot for regulatory purposes. The definitive assessment of Lirentelimab in EGIDs was that, despite its undeniable and potent biological activity, it failed to provide a demonstrable clinical benefit in large, well-controlled registrational studies.
Following the disappointing outcomes in EGIDs, Allakos Inc. pivoted its strategy in a high-risk attempt to salvage the Lirentelimab program by focusing on other inflammatory conditions where eosinophils and mast cells are strongly implicated: atopic dermatitis and chronic urticaria.[7] This strategic shift was supported by the drug's dual mechanism of action and some promising early-phase data. However, robust, placebo-controlled trials in these indications ultimately confirmed the same pattern of biological activity without clinical benefit, leading to the program's final termination.
Initial exploratory studies in other allergic conditions had provided a basis for optimism. A Phase 2a open-label, proof-of-concept study investigated Lirentelimab in 45 patients with various forms of chronic urticaria (CU) that were refractory to high-dose antihistamines.[16] The results were encouraging, showing marked improvements in disease control. For instance, in omalizumab-naive patients with chronic spontaneous urticaria (CSU), 92% achieved a complete response as measured by the Urticaria Control Test (UCT), and the mean Urticaria Activity Score (UAS7) was reduced by 73%.[16]
Separately, a Phase 1 trial (NCT03379311) was completed in patients with severe allergic eye diseases, including atopic keratoconjunctivitis, vernal keratoconjunctivitis, and perennial allergic conjunctivitis.[33] This study established the drug's tolerability in this population and provided preliminary signals of efficacy, further supporting the rationale to explore Lirentelimab's potential beyond EGIDs.[39]
The ATLAS trial (NCT05155085) was a randomized, double-blind, placebo-controlled Phase 2 study designed to evaluate the efficacy and safety of a subcutaneous formulation of Lirentelimab in 122 adult patients with moderate-to-severe atopic dermatitis inadequately controlled by topical medications.[8] The results, announced in January 2024, represented a significant blow to the program.[8]
The trial failed to meet its primary endpoint, which was the proportion of patients achieving at least a 75% improvement in the Eczema Area and Severity Index (EASI-75) at 14 weeks. The response rate in the Lirentelimab-treated group was only 23%, which was not statistically superior to the 18% response rate observed in the placebo group.[11] The study also missed its secondary endpoints. Consistent with previous trials, Lirentelimab demonstrated potent biological activity, with blood eosinophil counts decreasing by 96% in the treatment arm versus only 15% in the placebo arm.[8] This result reinforced the emerging theme: profound eosinophil depletion did not translate into clinical improvement.
Concurrently with the ATLAS results, Allakos announced the failure of the MAVERICK trial, a Phase 2b randomized, double-blind, placebo-controlled study in 123 adult patients with moderate-to-severe CSU refractory to antihistamines.[8] This trial was designed to build upon the promising open-label data with a rigorous, controlled study.
The outcome was definitive and disappointing. The trial missed its primary endpoint, which was the absolute change from baseline in the weekly Urticaria Activity Score (UAS7) at 12 weeks. The reduction in disease activity was virtually identical between the two groups, with a 27% reduction for patients on Lirentelimab and a 26% reduction for patients on placebo.[11] Once again, the drug's pharmacodynamic effect was confirmed, with a 95% reduction in blood eosinophils in the Lirentelimab group, but this had no bearing on the clinical outcome.[8]
The comprehensive failures of both the ATLAS and MAVERICK trials, where the drug performed no better than placebo, were even more conclusive than the mixed results in EGIDs. They solidified the conclusion that the issue was not specific to a particular organ system but was fundamental to the drug's therapeutic mechanism in humans. The potent biological effect of eosinophil depletion was clinically inert across multiple distinct inflammatory diseases, leaving the company with no viable path forward for Lirentelimab.
| Trial | Indication | Primary Endpoint | Lirentelimab Arm | Placebo Arm | Result |
|---|---|---|---|---|---|
| ATLAS (Phase 2) | Atopic Dermatitis | Proportion of patients achieving EASI-75 at 14 weeks | 23% | 18% | Missed Endpoint |
| MAVERICK (Phase 2b) | Chronic Spontaneous Urticaria | Absolute change from baseline in UAS7 at 12 weeks | -27% | -26% | Missed Endpoint |
| Data sourced from.8 | |||||
While the development of Lirentelimab was ultimately focused on larger indications like EGIDs and atopic dermatitis, the first-in-human clinical trial was conducted in patients with Indolent Systemic Mastocytosis (ISM), a rare disease primarily driven by the clonal proliferation and activation of mast cells.[6] The results from this early study provided the first clinical evidence of Lirentelimab's activity and tolerability, particularly highlighting the potential of its mast cell-inhibiting mechanism.
The Phase 1 trial (NCT02808793) was a single-ascending dose and multi-dose study conducted in Germany to evaluate the safety, tolerability, and preliminary efficacy of Lirentelimab in 25 adult patients with ISM who had an unsatisfactory response to available treatments.[31] The primary endpoint of the study was safety and tolerability, which was successfully met. The drug was found to be generally well tolerated, with no serious adverse events reported. The most common treatment-related adverse events were mild-to-moderate infusion-related reactions, including feeling hot (76%) and headache (48%).[32]
The study's secondary endpoints assessed changes in patient-reported symptoms and quality of life using validated, disease-specific instruments: the Mastocytosis Symptom Questionnaire (MSQ), the Mastocytosis Activity Score (MAS), and the Mastocytosis Quality of Life Questionnaire (MC-QoL).[31]
In the multi-dose cohort (Part C), patients treated with Lirentelimab for six months showed substantial and clinically meaningful improvements across all measured domains.[31] Median symptom severity scores improved significantly compared to baseline:
Furthermore, median MC-QoL scores improved across all domains, including a 57% improvement in the emotions domain, a 42% improvement in social life/functioning, and a 39% improvement in the symptoms domain.[32] These results were highly promising, suggesting that Lirentelimab's mast cell-inhibiting activity could translate into tangible benefits for patients in a disease where mast cells are the primary pathogenic driver. Despite these positive findings, Allakos ultimately prioritized the development of Lirentelimab for the larger EGID and dermatologic indications, and further investigation in the rare disease of ISM was not pursued.
Across its extensive clinical development program, spanning multiple diseases, patient populations, and formulations, Lirentelimab consistently demonstrated a manageable safety and tolerability profile. Safety concerns were not a contributing factor to the discontinuation of its development; rather, the decision was based entirely on a lack of efficacy.[7]
The overall safety database for Lirentelimab showed that the drug was generally well-tolerated.[40] The most frequently reported adverse events, aside from infusion-related reactions, were typically mild to moderate in severity and included nasopharyngitis (21% in a CU study) and headache (19% in a CU study, 48% in the ISM study).[16] Importantly, no new or unexpected safety signals emerged during the later-stage, larger clinical trials.[7] The incidence of serious adverse events was generally low and comparable between Lirentelimab and placebo groups in controlled studies.[42]
The most common and consistent treatment-related adverse event associated with Lirentelimab was the occurrence of infusion-related reactions (IRRs) or injection-related reactions (for the subcutaneous formulation).[1] These reactions were consistently reported at a higher incidence in patients receiving Lirentelimab compared to placebo across all major trials.
These IRRs were characterized as typically mild-to-moderate and transient, with symptoms including flushing, a feeling of warmth, headache, nausea, and/or dizziness.[9] They tended to occur primarily following the first infusion of the drug.[1] The incidence of these reactions varied across studies but was consistently elevated in the active treatment arms:
While more frequent with Lirentelimab, these reactions were considered manageable within the context of clinical trials and did not pose a significant barrier to the drug's development.
| Adverse Event | Trial / Indication | Lirentelimab Incidence (%) | Placebo Incidence (%) |
|---|---|---|---|
| Infusion/Injection-Related Reactions | ENIGMA 2 (EG/EoD, IV) | 34% | 14% |
| KRYPTOS (EoE, IV) | 39% (High Dose) 26% (Low Dose) | 12% | |
| ATLAS (AD, SC) | 18.5% | 6.2% | |
| MAVERICK (CSU, SC) | 18.2% | 8.2% | |
| Phase 2a CU (IV) | 43% | N/A (Open-label) | |
| Headache | Phase 1 ISM (IV) | 48% | N/A (Open-label) |
| Phase 2a CU (IV) | 19% | N/A (Open-label) | |
| Feeling Hot | Phase 1 ISM (IV) | 76% | N/A (Open-label) |
| Data sourced from.8 | |||
To support the development of a more convenient subcutaneous (SC) formulation for use in chronic diseases, Allakos conducted a Phase 1 study (NCT04324268) in adult healthy volunteers.[43] This trial was designed to evaluate the safety, tolerability, pharmacokinetics (PK), and bioavailability of the SC formulation relative to the established intravenous (IV) formulation.[43] The study assessed key PK parameters such as the area under the serum concentration-time curve (AUC) and pharmacodynamic effects, measured by changes in peripheral blood eosinophil counts, over an 85-day period.[43] While the specific numerical results of this study have not been publicly detailed, the subsequent use of the SC formulation in the large Phase 2 ATLAS and MAVERICK trials indicates that the study successfully established a viable dosing regimen and acceptable bioavailability for the SC route of administration.[8]
The story of Lirentelimab culminated in a definitive and conclusive end, providing a stark lesson in the challenges of translating a potent biological mechanism into clinical success. The decision to terminate the program was a direct consequence of repeated efficacy failures, prompting a critical re-examination of the underlying scientific hypotheses and reshaping the corporate strategy of its developer.
On January 16, 2024, Allakos Inc. issued a press release announcing the topline data from the Phase 2 ATLAS trial in atopic dermatitis and the Phase 2b MAVERICK trial in chronic spontaneous urticaria.[8] The announcement stated unequivocally that neither trial had met its primary endpoint. In light of these comprehensive failures, which followed the earlier pivotal trial disappointments in EGIDs, the company made the strategic decision to halt all further clinical development of Lirentelimab.[8] This decision marked the official end of a program that had once been the company's lead asset and a source of significant optimism.
The central question arising from the Lirentelimab program is why a drug that was exceptionally effective at its biological function—depleting eosinophils—failed to improve how patients felt. The consistent disconnect between histologic efficacy and symptomatic benefit across multiple diseases points away from trial-specific flaws and towards a more fundamental issue with the therapeutic hypothesis. Several non-mutually exclusive hypotheses can be considered:
The failure of Lirentelimab does not necessarily invalidate Siglec-8 as a therapeutic target. The highly selective expression on eosinophils and mast cells remains an attractive feature. However, the results suggest that a simple depleting and inhibiting monoclonal antibody may not be the optimal therapeutic modality. Future approaches targeting Siglec-8 may need to be more potent or employ different mechanisms. For example, antibody-drug conjugates (ADCs) could use a Siglec-8 antibody to deliver a highly potent cytotoxic payload directly to eosinophils and mast cells, ensuring more complete and rapid elimination. Alternatively, bispecific antibodies could be designed to engage Siglec-8 with one arm and another inhibitory receptor with the other to achieve deeper cellular inhibition.[39]
The clinical trial failures had a devastating impact on Allakos Inc. The announcement of the ENIGMA 2 and KRYPTOS results in December 2021 caused the company's stock to plummet by as much as 88%.[10] The final termination of the program in January 2024 precipitated another steep decline in valuation and forced a major corporate restructuring, which included laying off approximately 50% of its workforce to conserve capital.[11]
In the wake of Lirentelimab's failure, Allakos pivoted its focus and remaining resources to its next-generation candidate, AK006.[8] AK006 is a monoclonal antibody targeting Siglec-6, an inhibitory receptor expressed selectively on mast cells, but not eosinophils. The company's rationale was that AK006 appeared to provide deeper and broader mast cell inhibition than Lirentelimab in preclinical studies and might succeed where Lirentelimab had failed.[3] However, this strategic pivot also ended in failure. In 2024, Allakos announced that AK006 failed to show benefit in a Phase 1 trial for chronic spontaneous urticaria, leading to the discontinuation of that program and the elimination of another three-quarters of its remaining workforce.[44]
The clinical development of Lirentelimab offers a profound and cautionary narrative in the field of immunology and drug development. It highlights the significant gap that can exist between a compelling scientific rationale, potent biological activity, and the ultimate goal of achieving clinically meaningful patient benefit.
Lirentelimab was born from an elegant scientific concept: the selective targeting of the Siglec-8 receptor to simultaneously eliminate pathogenic eosinophils and inhibit inflammatory mast cells. Supported by robust preclinical data, the drug entered clinical trials with high expectations. Its pharmacodynamic effect was never in doubt; across every major trial in every indication, Lirentelimab proved to be exceptionally effective at depleting eosinophils from both blood and tissue.
However, this biological potency proved to be a clinical mirage. After a single promising Phase 2 result, the drug repeatedly failed in large, well-controlled studies to show that this eosinophil depletion translated into an improvement in patient symptoms. The stark disconnect between histologic resolution and patient-reported outcomes in EGIDs, followed by outright failures in atopic dermatitis and chronic spontaneous urticaria, demonstrated that the drug's powerful mechanism was, for reasons that are still being debated, clinically inert. Its journey serves as a powerful reminder that even the most well-designed, targeted therapies can fail if the underlying assumptions about disease pathophysiology are incomplete.
The Lirentelimab story leaves the scientific and medical community with critical lessons and unanswered questions.
In conclusion, while Lirentelimab did not become the transformative therapy it was hoped to be, its development was not without value. The rigorous clinical trials conducted have generated a wealth of data that challenges long-held beliefs about the pathophysiology of eosinophilic diseases. It stands as a crucial, albeit costly, scientific experiment that will undoubtedly shape the direction of research and drug development in immunology for years to come, guiding future efforts toward a more nuanced understanding of the complex interplay between inflammation, tissue pathology, and patient experience.
Published at: October 12, 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.