Dazodalibep is an investigational, second-generation therapeutic agent representing a significant potential advancement in the treatment of autoimmune diseases, most notably Sjögren's Syndrome. Classified as a non-antibody fusion protein, Dazodalibep functions as a highly specific antagonist of the CD40 ligand (CD40L), a critical component of the immune system's co-stimulatory axis. Its molecular architecture was strategically engineered to circumvent the severe thromboembolic toxicities that halted the development of first-generation anti-CD40L monoclonal antibodies, a feat achieved by eliminating the problematic Fc region. The drug's mechanism of action is centered on the blockade of the CD40/CD40L interaction, thereby disrupting the pathological communication between T cells, B cells, and other antigen-presenting cells that drives autoimmunity.
The clinical development program, now under the stewardship of Amgen following its acquisition of Horizon Therapeutics, is anchored by compelling results from a robust Phase 2, randomized, placebo-controlled, crossover trial (NCT04129164) in Sjögren's Syndrome. This study uniquely targeted two distinct patient populations: one with moderate-to-severe systemic disease activity and another with a high burden of patient-reported symptoms. Dazodalibep met its primary endpoints with high statistical significance in both groups, demonstrating a profound ability to reduce objective measures of systemic disease activity (measured by the ESSDAI score) and to provide clinically meaningful improvements in the debilitating symptoms of dryness, fatigue, and pain (measured by the ESSPRI score). The drug was found to be generally safe and well-tolerated, with no evidence of the thromboembolic events that plagued its predecessors.
These promising efficacy and safety data have prompted the initiation of a comprehensive pivotal Phase 3 program, which is currently underway and expected to yield results in 2026. Beyond Sjögren's Syndrome, Dazodalibep has also shown positive results in a Phase 2 study for Rheumatoid Arthritis and holds mechanistic promise for a range of other autoimmune conditions, including Systemic Lupus Erythematosus and Focal Segmental Glomerulosclerosis. Given the profound unmet medical need in Sjögren's Syndrome, for which no disease-modifying therapies are currently approved, Dazodalibep is poised to potentially become a first-in-class, paradigm-shifting treatment, establishing a new standard of care and validating the CD40/CD40L pathway as a key therapeutic target in modern immunology.
Dazodalibep (also known by the synonyms HZN-4920, MEDI-4920, VIB 4920, and AMG 611) is a biologic therapeutic agent classified as a novel, non-antibody fusion protein.[1] Unlike conventional monoclonal antibodies, its structure is distinct. It is described as a recombinant human tenascin C CD40 ligand-binding third fibronectin type III domain fusion protein.[6] This complex structure is engineered to have two primary functional domains: one side is designed to bind to serum albumin, while the other specifically targets and binds to the CD40 ligand (CD40L, also known as CD154).[7] This dual-binding characteristic is fundamental to its pharmacological properties, including its extended half-life and targeted biological activity.
The development of Dazodalibep was primarily led by Horizon Therapeutics, a biopharmaceutical company with a dedicated focus on developing treatments for rare, autoimmune, and severe inflammatory diseases.[7] The promising results generated under Horizon's stewardship, particularly in Sjögren's Syndrome, attracted significant industry attention. In 2024, the program was acquired by Amgen, a global biotechnology leader, as part of a larger corporate acquisition.[9] Amgen has since assumed responsibility for the drug's continued development and is the official sponsor of the ongoing pivotal Phase 3 clinical trials.[5] This transition to a major pharmaceutical entity underscores the high level of confidence in Dazodalibep's scientific and commercial potential.
The therapeutic potential of inhibiting the CD40/CD40L pathway has been recognized for decades. However, the clinical development of first-generation anti-CD40L monoclonal antibodies (mAbs) was uniformly terminated due to the emergence of severe and unacceptable thromboembolic complications, including deep vein thrombosis, pulmonary embolism, and stroke.[2] Subsequent investigations revealed that these adverse events were not a direct result of CD40L blockade itself but were an off-target effect mediated by the constant fragment (Fc) region of the antibody structure. The Fc region of these early mAbs inadvertently interacted with Fc$\gamma$RIIA receptors on platelets, causing platelet activation and aggregation, which led to the formation of blood clots.[2]
This understanding of the underlying mechanism of toxicity provided a clear path forward for a second generation of safer inhibitors. Dazodalibep was strategically and intelligently designed to overcome this critical liability. Its antigen-binding domains, which are responsible for neutralizing CD40L, were engineered onto a Tn3 scaffold, a non-mAb protein platform that completely lacks an Fc region.[2] This elegant feat of protein engineering effectively decouples the desired therapeutic effect (CD40L antagonism) from the source of the toxicity (Fc-mediated platelet activation). The success of this design has been validated both preclinically and clinically.
In vitro studies have confirmed that Dazodalibep does not induce platelet aggregation, and to date, the extensive clinical trial program involving healthy volunteers and patients with Sjögren's Syndrome and Rheumatoid Arthritis has not revealed any thromboembolic safety signals.[2] This successful de-risking of a highly promising but historically challenging biological target represents a significant achievement in drug development and has revived the CD40/CD40L pathway as a viable and compelling target for autoimmune therapy.
The interaction between the CD40 receptor and its cognate ligand, CD40L, is a cornerstone of the adaptive immune response. This signaling pathway functions as a critical co-stimulatory signal, essential for the full activation and function of multiple immune cell types.[2] The CD40 receptor is broadly expressed on the surface of antigen-presenting cells (APCs), such as B lymphocytes, macrophages, and dendritic cells, as well as non-hematopoietic cells like epithelial cells.[2] CD40L, its binding partner, is transiently expressed primarily on the surface of activated CD4+ T cells.[8]
In a healthy immune response, the engagement of CD40L on a T cell with CD40 on a B cell provides a necessary "second signal" (following the initial T-cell receptor signal) that licenses the B cell to undergo proliferation, affinity maturation, and immunoglobulin class-switching, ultimately leading to the production of high-affinity antibodies. In autoimmune diseases such as Sjögren's Syndrome, this pathway becomes dysregulated and chronically overactivated.[7] Pathological evidence confirms that the expression of both CD40 and CD40L is significantly upregulated within the inflamed salivary gland tissues of patients with Sjögren's, perpetuating a cycle of unchecked immune activation and tissue damage.[2]
Dazodalibep is designed to function as a potent and specific antagonist of CD40L.[2] By binding directly to the CD40L protein on the surface of activated T cells, Dazodalibep physically obstructs its ability to engage with the CD40 receptor on B cells and other APCs.[2] This action effectively severs a critical line of communication—or "immune chitchat"—between these key cellular players in the autoimmune response.[7]
This blockade of the co-stimulatory signal prevents the downstream activation cascades that are essential for the propagation of the autoimmune attack. The intervention is not broadly immunosuppressive in the manner of corticosteroids or traditional disease-modifying anti-rheumatic drugs (DMARDs). Instead, it is a highly targeted immunomodulatory approach. By precisely targeting a specific, pathological activation loop, Dazodalibep aims to restore a degree of immune homeostasis rather than simply inducing a global suppression of immune function. It is this precision that underpins its potential to be a true disease-modifying agent, as it addresses one of the fundamental mechanisms driving the disease process itself.
The downstream consequences of Dazodalibep's mechanism of action are profound and directly counter the key pathological hallmarks of Sjögren's Syndrome and other B-cell-mediated autoimmune diseases. The CD40/CD40L interaction is indispensable for the formation and maintenance of germinal centers (GCs)—specialized microanatomical structures within secondary lymphoid organs and, pathologically, in inflamed tissues (ectopic GCs).[2] It is within these GCs that B cells undergo the processes of somatic hypermutation and class-switch recombination to produce high-affinity, pathogenic autoantibodies.
By disrupting this pathway, Dazodalibep is designed to dismantle the cellular machinery responsible for autoantibody production. This leads to a reduction in the development of pathogenic B cells and their differentiation into autoantibody-secreting plasma cells.[15] Furthermore, the CD40/CD40L signaling axis is known to stimulate the production of a host of pro-inflammatory cytokines and chemokines, including interferon-alpha (IFN$\alpha$), tumor necrosis factor-alpha (TNF$\alpha$), interleukin-6 (IL-6), and CXCL13, a chemokine crucial for recruiting B cells and T cells into GCs.[2] The blockade of this pathway by Dazodalibep is therefore expected to have a broad anti-inflammatory effect by dampening the production of these key mediators of tissue damage.
The cornerstone of the Dazodalibep development program is the data from a large, multicenter Phase 2 clinical trial (NCT04129164), the results of which were published in Nature Medicine and presented at major rheumatology congresses.[7] The trial's design was particularly robust, incorporating the gold-standard features of randomization, double-blinding, and placebo control to minimize bias.[2]
A key innovation of the study was its crossover design. After an initial 24-week treatment period (Stage 1), participants who were initially randomized to receive Dazodalibep were switched to placebo for the subsequent 16 weeks (Stage 2), while those who had received placebo were switched to active treatment.[18] This methodology is powerful because it allows each participant to serve as their own control, increasing statistical efficiency and providing unambiguous evidence of the drug's effects upon initiation and withdrawal. The dosing regimen consisted of 1,500 mg of Dazodalibep administered intravenously, starting with three doses every two weeks followed by maintenance doses every four weeks.[22]
Recognizing the clinical heterogeneity of Sjögren's Syndrome, the trial was intelligently designed to evaluate Dazodalibep in two separate and well-defined patient populations.[7] This dual-population approach, which has been carried forward into the Phase 3 program, represents a sophisticated strategy to demonstrate efficacy across the full spectrum of the disease and sets a new standard for clinical trial design in complex autoimmune conditions.
Dazodalibep successfully achieved its primary endpoint with high statistical significance in both patient populations, a remarkable outcome that demonstrates its broad therapeutic effect on both the objective signs and subjective symptoms of Sjögren's Syndrome.
In Population 1, treatment with Dazodalibep resulted in a statistically significant and clinically meaningful reduction in systemic disease activity. At Day 169, the least-squares (LS) mean change from baseline in the total ESSDAI score was -6.3 for the Dazodalibep group, compared to -4.1 for the placebo group. This 2.2-point difference was highly significant (p=0.0167), indicating a substantial impact on the underlying organ-based disease activity.[15]
In Population 2, Dazodalibep demonstrated a profound effect on the symptoms that most impact patients' quality of life. The LS mean change from baseline in the total ESSPRI score at Day 169 was -1.80 for patients receiving Dazodalibep, versus only -0.53 for those on placebo. This difference of -1.27 was also highly statistically significant (p=0.0002).[15]
The improvement was consistent across all three domains of the ESSPRI score, with significant differences favoring Dazodalibep for dryness (p=0.0066), fatigue (p=0.0022), and pain (p=0.0010).[22] Furthermore, a significantly greater proportion of patients treated with Dazodalibep achieved a clinically meaningful response, defined as at least a 1-point or a 15% reduction in their ESSPRI score, compared to placebo (66.7% vs. 32.7%;
p=0.008).[22]
The data from the crossover stage of the trial provided compelling evidence for the drug's potent and durable biological effect. Participants who switched from placebo to Dazodalibep at Day 169 experienced a marked improvement in their disease scores by Day 365, with the ESSDAI score reduction deepening from -4.1 to -6.3 and the ESSPRI score reduction improving from -0.5 to -1.3.[18] Conversely, and perhaps more importantly, participants who had been treated with Dazodalibep and then switched to placebo largely maintained the clinical benefits they had achieved. For example, the 1.8-point reduction in ESSPRI score seen at Day 169 was sustained at a 1.9-point reduction by Day 365, despite being off the active drug for several months.[18] This sustained response suggests that Dazodalibep induces a durable modulation of the immune system, a key characteristic of a true disease-modifying therapy.
Table 1: Summary of Pivotal Phase 2 Efficacy Results in Sjögren's Syndrome (NCT04129164) at Day 169 |
---|
Patient Population |
Population 1 (ESSDAI ≥ 5) |
Population 2 (ESSPRI ≥ 5) |
Population 2 (ESSPRI ≥ 5) |
Population 2 (ESSPRI ≥ 5) |
Population 2 (ESSPRI ≥ 5) |
Population 2 (ESSPRI ≥ 5) |
Data compiled from sources.15 |
Across the clinical development program, Dazodalibep has consistently demonstrated an acceptable safety and tolerability profile.[2]
The incidence of treatment-emergent adverse events (TEAEs) was generally balanced between the Dazodalibep and placebo arms of the Phase 2 Sjögren's trial. Through Day 169, 68.5% of participants in the Dazodalibep group and 69.1% in the placebo group reported at least one TEAE, the vast majority of which were mild to moderate in severity.[16] The most common adverse events reported more frequently in the Dazodalibep arm were consistent with those expected from an immunomodulatory agent and included infections and gastrointestinal effects.
Table 3: Phase 2 Safety Profile of Dazodalibep in Sjögren's Syndrome (Most Frequent TEAEs through Day 169) |
---|
Adverse Event (MedDRA Preferred Term) |
COVID-19 |
Nasopharyngitis |
Anemia |
Diarrhea |
Headache |
Upper Respiratory Tract Infection |
Arthralgia |
Constipation |
Urinary Tract Infection |
Data compiled from sources.15 Specific percentages for all AEs were not consistently provided across all sources, but these were listed as the most frequent. |
A critical finding from the safety analysis is the absence of any signal for thromboembolic events, the very toxicity that doomed the first generation of anti-CD40L therapies.[2] This favorable safety profile validates the strategic molecular engineering of Dazodalibep and is paramount to its viability as a long-term therapy for chronic autoimmune diseases.
The incidence of serious adverse events was low and comparable between treatment groups. In the Sjögren's trial, three SAEs were reported in the Dazodalibep group (pneumonia influenza, post-acute COVID-19 syndrome, and gammopathy) and one in the placebo group (neutropenia).[22] Importantly, all of these SAEs were deemed by the study investigators to be unrelated to the investigational drug.[16]
The clinical efficacy of Dazodalibep was further substantiated by a robust set of biomarker data that provides direct evidence of its intended mechanism of action in vivo.[17] Treatment with Dazodalibep led to rapid and significant reductions in a range of blood biomarkers indicative of T- and B-cell co-stimulation.
Flow cytometry analysis of peripheral blood mononuclear cells revealed that Dazodalibep treatment significantly reduced the populations of key pathogenic immune cells. These included proliferating memory B cells (Ki67+CD27+), plasmablasts (CD27highCD38high), which are immediate precursors to antibody-secreting cells, and T follicular helper cells, the specific T-cell subset responsible for providing help to B cells within germinal centers.[17] These findings confirm that Dazodalibep is effectively disrupting the B-cell activation and differentiation pathway at a cellular level.
Consistent with its impact on germinal center activity, Dazodalibep treatment led to a significant decrease in the serum levels of CXCL13, a chemokine that is essential for the organization of GCs and is considered a reliable surrogate marker of GC activity.[17] Reductions in autoantibody levels, such as Rheumatoid Factor (RF), were also observed.[17] A comprehensive serum proteomics analysis further identified a signature of 29 proteins associated with B-cell, T-cell, and dendritic cell activation that were elevated in Sjögren's patients and were significantly reduced following treatment with Dazodalibep.[20] Together, these biomarker data provide a clear mechanistic link between the drug's action on the CD40/CD40L pathway and the clinical improvements observed in patients.
Building on the strength of the Phase 2 results, Amgen has launched a comprehensive Phase 3 program designed to definitively establish the efficacy and safety of Dazodalibep and support applications for regulatory approval.[7] The program mirrors the successful dual-population strategy of the Phase 2 trial.
Table 2: Overview of the Dazodalibep Phase 3 Program |
---|
Study Identifier (NCT Number) |
NCT06104124 |
NCT06245408 |
NCT06747949 |
Data compiled from sources.4 |
The central role of the CD40/CD40L pathway in a wide range of immune-mediated diseases suggests that Dazodalibep's therapeutic utility may extend far beyond Sjögren's Syndrome. The consistent biomarker effects observed across different disease states indicate that the drug is targeting a fundamental pathological mechanism, positioning it as a potential "pipeline in a product" with broad applicability.
Dazodalibep has also been evaluated in a Phase 2, placebo-controlled clinical trial (MIDORA) in patients with moderate-to-severe active Rheumatoid Arthritis (RA) who had an inadequate response to conventional therapies.[16] The trial met its primary endpoint, with Dazodalibep demonstrating statistically significant improvements in the Disease Activity Score-28 with C-Reactive Protein (DAS28-CRP) compared to placebo. Significant benefits were also observed in other measures of disease activity, including tender and swollen joint counts.[19] The biomarker profile in the RA trial was remarkably consistent with that seen in the Sjögren's study, showing reductions in plasmablasts, T follicular helper cells, and serum levels of CXCL13 and RF.[19] These positive results provide strong clinical proof-of-concept for Dazodalibep's efficacy in a second major autoimmune indication.
The strong mechanistic rationale for CD40L blockade has prompted investigation into several other conditions. The pathway is known to be heavily implicated in the pathogenesis of Systemic Lupus Erythematosus (SLE) and lupus nephritis, making these high-priority areas for potential future development.[1]
Amgen has also announced plans to investigate Dazodalibep for the treatment of Focal Segmental Glomerulosclerosis (FSGS), a rare and serious kidney disease characterized by scarring of the kidney's filtering units.[18] In addition, the field of organ transplantation is actively exploring next-generation costimulation blockers to replace or reduce the need for calcineurin inhibitors, which are effective but carry long-term toxicity risks. CD40L antagonists, including Dazodalibep, have shown promising efficacy and safety in preclinical and early clinical studies for the prevention of kidney transplant rejection.[1]
The therapeutic landscape for Sjögren's Syndrome is characterized by a profound unmet medical need. There are currently no therapies approved by the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA) that are capable of modifying the underlying course of the disease.[2] This treatment vacuum forces clinicians and patients to rely on a patchwork of therapies that are either purely symptomatic or are broad-acting immunosuppressants used off-label with limited and inconsistent evidence of efficacy.
The current standard of care is largely palliative.[2] Dry eyes and dry mouth are managed with a variety of over-the-counter products like artificial tears and saliva substitutes, as well as prescription medications known as secretagogues (e.g., pilocarpine, cevimeline) that stimulate glandular secretion.[32] For patients with systemic manifestations such as arthritis, vasculitis, or organ involvement, clinicians resort to off-label use of drugs approved for other rheumatic diseases. These include hydroxychloroquine (Plaquenil), corticosteroids, and other DMARDs like methotrexate.[31] Biologics that target B cells, such as rituximab, are also used off-label for severe disease, but large, well-controlled clinical trials like TRACTISS and TEARS failed to demonstrate a significant benefit on the primary endpoints, leaving their role in treatment uncertain.[31]
Against this backdrop, Dazodalibep's potential is transformative. Its demonstrated ability to significantly improve both systemic disease activity (ESSDAI) and patient-reported symptoms (ESSPRI) positions it to become the first-ever approved disease-modifying therapy for Sjögren's Syndrome.[7]
Dazodalibep remains an investigational product and has not yet been submitted for marketing approval to any regulatory agency.[18] The ongoing Phase 3 trials are designed as registrational studies, meaning their results will form the core of the data package submitted to agencies like the FDA and EMA. The meticulous design of the clinical program, particularly the successful demonstration of efficacy in two distinct, pre-specified patient populations, is likely to be viewed very favorably by regulators.
Should Dazodalibep gain approval, its commercial opportunity is substantial. As a first-to-market, disease-modifying therapy in an area of complete unmet need, it would face no direct competition at launch. This would allow for rapid physician adoption and significant market penetration. The efficacy demonstrated across both systemic and symptomatic patient populations could support a broad label, making the drug a potential treatment option for a large proportion of the moderate-to-severe Sjögren's patient population. The availability of a truly effective therapy would also likely drive an increase in diagnosis rates, as both patients and physicians would be more motivated to seek a definitive diagnosis based on established criteria, such as the 2016 ACR/EULAR Classification Criteria [40], knowing that a meaningful intervention is available.
Dazodalibep stands out as a highly promising therapeutic candidate, distinguished by its intelligent molecular design, a clear and validated mechanism of action, and a robust body of clinical evidence. The successful circumvention of the thromboembolic risks that plagued earlier CD40L inhibitors is a landmark achievement in protein engineering and has reopened a pivotal pathway in immunology for therapeutic intervention.
The Phase 2 clinical trial in Sjögren's Syndrome was exceptionally well-designed and yielded impressively positive results. The demonstration of statistically significant and clinically meaningful efficacy on both objective systemic endpoints and subjective patient-reported outcomes across two distinct disease phenotypes is a rare and powerful finding. This, combined with a favorable safety profile and compelling biomarker data that corroborates the drug's mechanism in vivo, provides a very strong foundation for the ongoing pivotal Phase 3 program.
If the results of the HZNP-DAZ-301 and HZNP-DAZ-303 trials replicate the findings from Phase 2, Dazodalibep is on a clear trajectory to become the first-in-class, disease-modifying standard of care for patients with moderate-to-severe Sjögren's Syndrome. Such an approval would not only revolutionize the management of this debilitating disease but would also firmly establish the CD40/CD40L axis as a validated, druggable target in rheumatology. The positive data in Rheumatoid Arthritis further suggests that Dazodalibep has the potential to become a valuable platform therapy for a multitude of B-cell-driven autoimmune diseases. The key inflection points for the program will be the topline data readouts from the ongoing Phase 3 studies, which are anticipated in 2026 and will be critical in determining the ultimate regulatory and clinical fate of this important new medicine.
Published at: September 29, 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.