Biotech
2639474-65-8
Dazukibart is an investigational biologic therapeutic agent, distinguished as a mouse-derived, humanized IgG1κ monoclonal antibody.[1] Its primary therapeutic classification is an anti-interferon beta (IFNβ) therapy, indicating its mechanism is centered on neutralizing the activity of this specific cytokine.[2] The development of Dazukibart is principally focused on addressing idiopathic inflammatory myopathies (IIM), with particular emphasis on dermatomyositis (DM) and polymyositis (PM).[2] Furthermore, its therapeutic potential has been explored in the context of lupus, encompassing both systemic lupus erythematosus (SLE) and cutaneous lupus erythematosus (CLE).[3]
The significance of Dazukibart in the landscape of emerging therapies stems from its targeted approach. It is designed to intervene in diseases where IFNβ is understood to be a key pathogenic driver, contributing to the underlying inflammation and immune dysregulation characteristic of these conditions.[5] This targeted strategy differentiates it from broader immunosuppressive agents and aligns with the contemporary paradigm of precision medicine in autoimmune disorders. The development program reflects a strategic focus on IFNβ-mediated diseases, suggesting an intent to leverage a well-defined mechanism of action across multiple autoimmune conditions where IFNβ is a common pathological denominator. This approach, if successful, could offer an efficient pathway to addressing unmet needs in several related disorders.
The precise identification of an investigational drug is paramount for accurate scientific communication and regulatory tracking. Dazukibart is known by several identifiers:
The designation of Dazukibart as a "humanized" monoclonal antibody is a critical aspect of its design. Therapeutic antibodies initially developed in murine systems can provoke an immune response in human recipients (known as a human anti-mouse antibody, or HAMA, response). This immunogenicity can diminish the drug's effectiveness over time and potentially lead to adverse reactions. The humanization process involves grafting the antigen-binding regions (complementarity-determining regions, or CDRs) from the original murine antibody onto a human antibody framework. This sophisticated bioengineering technique aims to preserve the specific IFNβ-targeting capability of the parent antibody while minimizing its potential to be recognized as foreign by the human immune system. This characteristic is particularly vital for a therapeutic agent intended for chronic or long-term administration, as is often required in the management of autoimmune diseases. The observation of anti-drug antibodies (ADAs) in a Phase 1 clinical trial underscores that even with humanization, careful monitoring for immunogenic responses remains an essential component of its clinical development.[15]
Table 1: Dazukibart - Key Identifiers and Properties
Property | Detail | Reference(s) |
---|---|---|
Generic Name | Dazukibart | 1 |
Synonyms/Code Names | PF-06823859, PF06823859 | 8 |
DrugBank ID | DB18429 | 8 |
CAS Number | 2639474-65-8 | 1 |
Drug Type | Biotech | 8 |
Molecular Class | Humanized IgG1κ monoclonal antibody | 1 |
Target | Interferon-beta (IFNβ) | 1 |
Molecular Weight | Approx. 145.36 kDa | 1 |
Developer | Pfizer Inc. | 9 |
Dazukibart is under development by Pfizer Inc., a global pharmaceutical corporation.[4] Pfizer's involvement signifies a substantial commitment of resources towards the research, clinical evaluation, and potential future commercialization of this therapeutic agent. The inclusion of Dazukibart in Pfizer's publicly disclosed product pipeline updates further confirms its ongoing strategic importance within their Inflammation and Immunology portfolio.[4]
Dazukibart is engineered as a potent and selective humanized IgG1 neutralizing monoclonal antibody that is specifically directed against interferon-beta (IFNβ).[1] Its therapeutic action is predicated on its ability to bind with high affinity to IFNβ, thereby inhibiting the biological activities of this cytokine.[5] The specificity for IFNβ, as opposed to a broader inhibition of all Type I interferons, represents a deliberate therapeutic strategy. While both IFNα and IFNβ are Type I interferons and utilize common signaling pathways, their primary cellular sources and potentially nuanced physiological and pathological roles may differ. For instance, plasmacytoid dendritic cells (pDCs) are major producers of IFN-α, whereas other cell types, including macrophages and fibroblasts, are primary synthesizers of IFN-β.[17] Targeting IFNβ alone might therefore offer a more refined immunomodulatory effect. This could potentially reduce the spectrum of immune suppression and associated adverse events (such as heightened susceptibility to certain viral infections that are often managed by IFNα activity) compared to a pan-Type I IFN inhibitor, while still maintaining efficacy in diseases where IFNβ is identified as a critical pathogenic driver, such as dermatomyositis.[7] This targeted specificity could be instrumental in optimizing the therapeutic window of Dazukibart.
The rationale for developing an anti-IFNβ therapy like Dazukibart is firmly rooted in the growing body of evidence implicating Type I interferons, and IFNβ specifically, in the pathogenesis of several autoimmune diseases.
Dazukibart exerts its therapeutic effect by functioning as a neutralizing antibody. It is designed to bind with high specificity and affinity to IFNβ, thereby physically preventing IFNβ from interacting with its cognate cell surface receptors, primarily the Type I interferon receptor (IFNAR) complex.1
By sequestering IFNβ and blocking its receptor binding, Dazukibart effectively interrupts the downstream intracellular signaling pathways that are normally activated by this cytokine. This blockade is anticipated to lead to a modulation of the overactive immune responses characteristic of the targeted autoimmune diseases. Consequently, this should result in a reduction in inflammation and an alleviation of disease symptoms in conditions such as DM, PM, and lupus.2
One source, a product page from ThermoFisher Scientific, describes Dazukibart's mechanism as "inhibiting the enzyme responsible for the synthesis of pro-inflammatory cytokines".23 While IFNβ itself is a pro-inflammatory cytokine, and its neutralization by Dazukibart would indeed lead to a decrease in the overall pro-inflammatory environment (including a reduction in other cytokines that are induced by IFNβ), the primary molecular mechanism of action for a monoclonal antibody like Dazukibart is direct binding and neutralization of its target protein (IFNβ). It does not act as an enzyme inhibitor in the classical pharmacological sense. The description provided in 23 and 24 likely refers to the ultimate downstream consequences of IFNβ blockade rather than its direct molecular interaction.
Dazukibart is characterized as a humanized IgG1κ monoclonal antibody.1 The IgG1 isotype is frequently employed for therapeutic antibodies due to its favorable pharmacokinetic properties and its capacity to mediate effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). However, for a neutralizing antibody like Dazukibart, the primary therapeutic effect is achieved through high-affinity binding to its target, IFNβ, thereby preventing its interaction with cellular receptors.
The antibody was initially derived from a murine (mouse) source and subsequently underwent a humanization process to reduce potential immunogenicity in human subjects.1 The approximate molecular weight of Dazukibart is 145.36 kDa, which is typical for a full-length IgG molecule.1 In terms of physical appearance, it is described as a liquid that is colorless to light yellow.1
The pharmacokinetic profile of Dazukibart has been evaluated in Phase 1 clinical studies involving healthy adult volunteers in China (Study 1, receiving a single 900 mg dose) and Japan (Study 2, receiving single doses of 300 mg or 900 mg).[15]
The pharmacodynamic effects of Dazukibart relate to its biological impact on the IFNβ pathway. A key objective in the clinical trial program has been to assess the "target engagement" of Dazukibart, confirming that the antibody effectively interacts with and neutralizes IFNβ in vivo.6
The fundamental mechanism of action – the neutralization of IFNβ – implies that the pharmacodynamic consequences would include a reduction in IFNβ-mediated signaling. This could manifest as a decrease in the expression of IFN-inducible genes, often referred to as the "IFN signature," which is a hallmark of IFN pathway activation in various autoimmune diseases. The Lancet publication detailing the Phase 2 trial in dermatomyositis explicitly mentions the assessment of target engagement as part of the study's aims.6
The potential for Dazukibart to elicit an immune response in patients (immunogenicity) has been evaluated in early clinical studies.
In the Phase 1 study conducted in China (Study 1), none of the participants developed anti-drug antibodies (ADAs) to Dazukibart.15
In contrast, in the Phase 1 study conducted in Japan (Study 2), 20.0% of participants tested positive for treatment-induced ADAs. Furthermore, these ADAs were found to be neutralizing antibodies (NAbs) in these individuals, meaning they had the potential to interfere with the drug's ability to bind to IFNβ.15
The development of ADAs, and particularly NAbs, is a critical consideration for all biologic therapies. ADAs can affect the drug's pharmacokinetics (e.g., by accelerating clearance), reduce its efficacy (by neutralizing its activity), and, in some cases, lead to safety concerns such as hypersensitivity reactions. The observed difference in ADA rates between the Chinese (0%) and Japanese (20%) cohorts in these small Phase 1 studies, while not necessarily statistically definitive due to limited sample sizes, highlights the importance of comprehensive immunogenicity assessment in larger, more diverse patient populations throughout the clinical development program. A 20% incidence of ADAs/NAbs could be clinically relevant if it translates to a loss of efficacy or safety issues in a subset of patients undergoing long-term treatment. This underscores the necessity for continuous and robust monitoring of immunogenicity in ongoing and future trials.
Dermatomyositis (DM) and polymyositis (PM) are chronic and often debilitating autoimmune disorders characterized by inflammation of the muscles, leading to progressive muscle weakness. DM is further distinguished by the presence of characteristic skin rashes.2 These conditions significantly impact patients' quality of life and functional abilities.
The current standard of care for DM and PM typically involves the use of systemic corticosteroids to control inflammation, often in conjunction with other immunosuppressive agents such as hydroxychloroquine, methotrexate, azathioprine, mycophenolate mofetil, or intravenous immunoglobulin (IVIG).5 Despite these available treatments, a substantial proportion of patients experience treatment-refractory disease, where symptoms persist or recur despite adequate trials of standard therapies. This highlights a significant unmet medical need for novel, more effective, and better-tolerated treatment options for individuals with DM and PM.5
Dazukibart has progressed through Phase 1 and Phase 2 clinical development and is currently in Phase 3 trials for DM and PM. Key studies in this program include:
The progression from promising Phase 2 results to large-scale Phase 3 trials underscores the commitment to thoroughly evaluate Dazukibart for these challenging conditions.
In clinical trials, Dazukibart has been consistently administered as an intravenous (IV) infusion. The infusion process typically takes approximately one hour.2
The dosing regimens explored in these trials include:
The Phase 2 trial (NCT03181893) provided key efficacy data, particularly for patients with skin-predominant DM:
Based on the Phase 2 trial (NCT03181893), Dazukibart was generally well tolerated.[6]
Long-term safety is a primary focus of ongoing extension studies (e.g., NCT06698796).[2] Key safety parameters being monitored in these long-term evaluations include laboratory abnormalities, changes in vital signs, electrocardiogram (ECG) abnormalities (with a particular focus on QTc prolongation, a measure of cardiac repolarization), and lung function assessments.[2]
Table 2: Summary of Key Clinical Trials for Dazukibart in Dermatomyositis and Polymyositis
Trial ID | Phase | Condition(s) | Key Objectives/Endpoints | Dazukibart Dose(s) | Key Efficacy Outcomes (Week 12, Pooled Skin FAS vs Placebo) | Salient Safety Findings | Status | Reference(s) |
---|---|---|---|---|---|---|---|---|
NCT03181893 | 2 | Dermatomyositis (skin-predominant, muscle-predominant) | Efficacy (CDASI-A change), Safety, Target Engagement | 150 mg IV Q4W, 600 mg IV Q4W | 600mg: CDASI-A change -19.2 (Placebo-adj. diff. -16.3, p<0.0001); 150mg: CDASI-A change -16.6 (Placebo-adj. diff. -13.7, p<0.0001) | TEAEs: ~80% all groups. Most common: infections. 1 death (HLH/MAS) in 600mg group during follow-up. | Completed | 5 |
NCT05895786 | 3 | Dermatomyositis (DM), Polymyositis (PM) | Efficacy, Safety | IV Q4W (dose likely based on Ph2) | Primary completion July 2026 | Ongoing monitoring | Recruiting | 4 |
NCT06698796 | 3 (OLE) | Dermatomyositis (DM), Polymyositis (PM) | Long-term safety and efficacy | IV (dose likely based on parent study) | Long-term outcomes | Long-term safety monitoring | Planned (Jan 2025) | 2 |
Systemic lupus erythematosus (SLE) is a complex chronic autoimmune disease that can affect multiple organ systems, while cutaneous lupus erythematosus (CLE) primarily manifests with skin involvement.[12] As detailed in Section 2.2, Type I interferons, including IFNβ, are strongly implicated in the pathophysiology of lupus. Many patients with SLE and CLE experience persistent skin symptoms that do not respond adequately to standard treatments, such as topical therapies, antimalarials, or systemic immunosuppressants, thus representing an area of unmet medical need.[12] The rationale for investigating an anti-IFNβ therapy like Dazukibart in lupus stems from the desire to specifically target this component of the IFN pathway.
Dazukibart is currently in Phase 2 of clinical development for the treatment of SLE and CLE.[3] The clinical program for lupus is at an earlier stage compared to that for DM and PM, reflecting a common pharmaceutical development strategy of prioritizing indications with the strongest initial evidence or perhaps a more clearly defined role for the specific target.
Consistent with its use in DM/PM trials, Dazukibart is administered via intravenous (IV) infusion in the NCT05879718 lupus trial.13
The dosing schedule in this trial involves administration every 4 weeks for the initial three doses (Day 1, Week 4, and Week 8). Subsequently, the dosing interval is extended to every 8 weeks, from Week 16 up to Week 40.12 This dosing regimen, particularly the extension of the interval in the later phase, may be designed to explore maintenance of effect with less frequent dosing after an initial induction period.
As the NCT05879718 trial is currently ongoing, specific efficacy results are not yet available in the provided information. The study is designed to assess whether Dazukibart can lead to improvements in skin manifestations, primarily measured by changes in the Cutaneous Lupus Erythematosus Disease Area and Severity Index activity (CLASI-A) score, as well as other systemic symptoms in participants with SLE.[12] Safety and tolerability will also be key outcomes rigorously evaluated throughout the trial.
Table 3: Summary of Key Clinical Trials for Dazukibart in Systemic/Cutaneous Lupus Erythematosus
Trial ID | Phase | Condition(s) | Key Objectives/Endpoints | Dazukibart Dose(s) | Key Efficacy Outcomes | Salient Safety Findings | Status | Reference(s) |
---|---|---|---|---|---|---|---|---|
NCT05879718 | 2 | Cutaneous Lupus Erythematosus (CLE), Systemic Lupus Erythematosus (SLE) with skin symptoms | Clinical effect (CLASI-A), PD, PK, Safety | IV Q4W then Q8W | Pending trial completion | Ongoing monitoring | Recruiting | 3 |
Dazukibart has received several important regulatory designations that acknowledge its potential to address unmet medical needs in rare and serious conditions. These designations can facilitate and expedite the drug development and review process.
The Orphan Drug status is a significant acknowledgment. It is granted to therapies intended for rare diseases or conditions (affecting fewer than 200,000 people in the U.S., or for the E.U., affecting not more than 5 in 10,000 people and being life-threatening or chronically debilitating). This status provides the developing company with various incentives, such as periods of market exclusivity upon approval, tax credits for clinical research, and waivers or reductions in regulatory fees, all designed to encourage the development of treatments for underserved patient populations.
In addition to Orphan Drug status, Dazukibart has benefited from other programs designed to accelerate its development:
The combination of Orphan Drug, Fast Track, and PRIME designations for Dazukibart in the context of Dermatomyositis signals a strong consensus among regulatory authorities regarding the severity and rarity of the condition, the substantial unmet medical need for more effective treatments, and the promising nature of the early data supporting Dazukibart's potential. Such comprehensive regulatory support can considerably shorten the timeline to potential market availability if the ongoing Phase 3 trials yield positive results.
Based on the available information, Dazukibart is an investigational medicine and has not yet received marketing approval from any regulatory agency in any jurisdiction.[2] It is actively being evaluated in Phase 2 and Phase 3 clinical trials. Pfizer's pipeline updates consistently list Dazukibart as a New Molecular Entity (NME) in Phase 3 development for DM and PM, and in Phase 2 development for Lupus.[4]
Table 4: Regulatory Designations for Dazukibart
Designation Type | Regulatory Agency | Indication | Reference(s) |
---|---|---|---|
Orphan Drug | FDA (U.S.) | Dermatomyositis | 4 |
Orphan Drug | EMA (E.U.) | Dermatomyositis | 4 |
Fast Track | FDA (U.S.) | Dermatomyositis | 4 |
PRIME | EMA (E.U.) | Dermatomyositis | 4 |
The provided research materials do not contain detailed results from specific preclinical efficacy or toxicology studies conducted directly with Dazukibart (PF-06823859) itself. Snippets [2] and [28], for instance, mention preclinical studies in a general context for anti-IFNβ therapies or other antibodies but do not offer data specific to Dazukibart.
However, the progression of Dazukibart into Phase 3 clinical trials inherently implies that a comprehensive package of preclinical data was generated and submitted to regulatory authorities (such as the FDA and EMA) to support the initiation of human studies. Standard drug development for a biologic like a monoclonal antibody involves an extensive preclinical phase. This typically includes:
While the specific outcomes of these Dazukibart-specific preclinical studies are not detailed in the available snippets, the robust scientific rationale derived from the known role of IFNβ in the target diseases serves as strong indirect preclinical support:
In essence, the development of Dazukibart has likely leveraged the extensive existing body of scientific research on the role of Type I interferons, and IFNβ specifically, in autoimmune diseases. This foundational knowledge, combined with Dazukibart-specific preclinical data (not available in the snippets), would have formed the necessary justification for advancing the drug into clinical trials. The absence of these specific preclinical results in the provided material represents a gap in the dataset for this report but does not negate the mandatory preclinical work that underpins progression to human testing.
Dazukibart, through its selective targeting and neutralization of interferon-beta, has demonstrated considerable promise as a novel therapeutic agent for autoimmune conditions, particularly Dermatomyositis (DM) and Polymyositis (PM), and is also under investigation for Lupus.[5] The positive results from the Phase 2 clinical trial in DM, especially the significant improvements observed in skin manifestations as measured by the CDASI-A score, are clinically meaningful and address a major component of the disease burden for these patients.[6] Should the ongoing Phase 3 trials corroborate these efficacy findings and establish a favorable long-term safety profile, Dazukibart could emerge as a valuable new addition to the therapeutic armamentarium. Its targeted mechanism offers a distinct advantage, particularly for patients who are refractory to, or intolerant of, existing standard-of-care treatments, which often involve broader immunosuppression.[5]
Current therapeutic strategies for DM and PM predominantly rely on corticosteroids and conventional disease-modifying antirheumatic drugs (DMARDs), which exert broad immunosuppressive effects.[5] While often effective in controlling acute inflammation, these agents are associated with a wide range of potential short-term and long-term side effects, limiting their utility, especially in chronic disease management. Dazukibart represents a more targeted immunomodulatory approach. By specifically neutralizing IFNβ, a cytokine identified as a key driver in the pathogenesis of these conditions, Dazukibart aims to interrupt disease processes with potentially greater precision. This specificity holds the promise of an improved benefit-risk profile compared to less targeted immunosuppressants. However, direct comparative clinical trial data between Dazukibart and existing therapies are not yet available from the provided information.
The development of Dazukibart directly addresses several critical unmet medical needs in the management of autoimmune myopathies. There is a pressing need for effective and well-tolerated therapies for patients with moderate-to-severe DM and PM who have not responded adequately to, or cannot tolerate, current standard treatments.[5] Dazukibart offers a novel mechanism of action that could provide benefit in this treatment-refractory population. Furthermore, if its efficacy is confirmed, it could serve as a corticosteroid-sparing agent, reducing the cumulative toxicity associated with long-term steroid use, or provide a viable alternative for patients for whom conventional immunosuppressants are contraindicated or have failed.
Pfizer is actively pursuing the clinical development of Dazukibart, with the compound advancing through Phase 3 trials for DM and PM (notably NCT05895786 and the planned open-label extension NCT06698796) and Phase 2 trials for Lupus (NCT05879718).[2] The consistent inclusion of Dazukibart in Pfizer's pipeline updates underscores its strategic value within their Inflammation and Immunology franchise.[4] The emphasis in clinical trials on assessing "target engagement" [6] suggests a scientifically driven approach to understand the drug's biological effects at a molecular level, which may also aid in the identification of biomarkers predictive of patient response. While Pfizer maintains a broad immunology portfolio, including Janus kinase (JAK) inhibitors [27], Dazukibart represents a distinct therapeutic modality as a biologic agent targeting a specific cytokine. This "pipeline in a product" strategy, investigating Dazukibart across multiple IFNβ-implicated autoimmune diseases, is a common approach for targeted therapies. Success in one indication can bolster the probability of success in others sharing similar underlying pathophysiology, thereby leveraging research and development investments. However, this strategy also carries the risk that challenges in one area (e.g., unforeseen safety signals or less-than-expected efficacy for a specific disease) could impact the broader program. The current phased development, with DM/PM more advanced than Lupus, likely reflects a prioritization based on the strength of the IFNβ link or the perceived unmet need.
Several challenges and limitations need to be considered as Dazukibart progresses through clinical development:
Dazukibart (PF-06823859) is an investigational humanized IgG1κ monoclonal antibody, developed by Pfizer, that selectively targets and neutralizes interferon-beta (IFNβ). This targeted approach positions Dazukibart as a potentially significant advancement in the treatment of autoimmune diseases where IFNβ is implicated as a key pathogenic mediator.
Clinical development has shown particular promise in Dermatomyositis (DM), with Phase 2 trial results, notably published in The Lancet [6], demonstrating statistically significant and clinically meaningful improvements in skin disease activity. These findings suggest that Dazukibart has the potential to address substantial unmet medical needs, especially for patients with moderate-to-severe DM who are refractory to or intolerant of current standard-of-care therapies. The drug has received Orphan Drug, Fast Track, and PRIME designations from regulatory authorities for DM, underscoring its perceived potential and the urgency for new treatments in this area.[4]
Ongoing Phase 3 trials in DM and Polymyositis (PM), alongside Phase 2 investigations in Systemic and Cutaneous Lupus Erythematosus, will be crucial in further defining Dazukibart's efficacy, long-term safety profile, and overall clinical utility across diverse patient populations. Continued rigorous monitoring for potential safety signals, such as the isolated case of HLH/MAS [6], and the potential impact of immunogenicity [15], will be essential components of its late-stage development and any subsequent post-marketing surveillance.
Dazukibart exemplifies the ongoing evolution in autoimmune disease treatment towards more targeted biologic therapies, moving beyond broad immunosuppression. If successfully developed and approved, Dazukibart could offer a novel, mechanism-based therapeutic option, potentially improving outcomes and quality of life for individuals burdened by these challenging autoimmune conditions. The strong scientific rationale, supported by the correlation of IFNβ with disease activity and the focus on target engagement, also hints at a future where such therapies might be utilized in a more personalized, biomarker-guided manner.
Published at: June 11, 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.