FDA Approve Twice-Yearly Depemokimab for Severe Eosinophilic Asthma Management

FDA Approve Twice-Yearly Depemokimab for Severe Eosinophilic Asthma Management

Twice-Yearly Depemokimab: A New Frontier in Severe Eosinophilic Asthma Management

Highlights

  • The US FDA has approved depemokimab-ulaa (Exdensur) as an add-on maintenance therapy for patients aged 12 and older with severe eosinophilic asthma.
  • As the first ultra-long-acting biologic targeting IL-5, depemokimab is administered once every six months (twice yearly).
  • Phase III SWIFT trials demonstrated a significant reduction in asthma exacerbations (up to 58%) compared to placebo.
  • The therapy addresses a critical unmet need for patients with type 2 inflammation who remain symptomatic despite high-dose inhaled corticosteroids.

Introduction: Addressing the Burden of Severe Eosinophilic Asthma

Severe asthma remains a significant clinical challenge, affecting approximately 5-10% of the total asthma population but accounting for a disproportionate share of healthcare utilization and costs. For patients with the eosinophilic phenotype, characterized by type 2 (T2) inflammation, the primary driver of airway pathology is the overproduction and activation of eosinophils. While existing monoclonal antibodies targeting interleukin-5 (IL-5) or its receptor have revolutionized care, the requirement for frequent dosing—typically every four to eight weeks—can pose a barrier to long-term adherence and patient quality of life.

The recent FDA approval of depemokimab-ulaa (Exdensur) marks a paradigm shift in the management of severe asthma. By leveraging an ultra-long-acting pharmacokinetic profile, depemokimab offers the potential for sustained eosinophil suppression with only two subcutaneous doses per year. This approval provides a vital new option for patients whose disease remains uncontrolled on standard-of-care inhaled therapies.

The Pharmacology of Depemokimab: An Ultra-Long-Acting IL-5 Inhibitor

Depemokimab is a humanized monoclonal antibody designed with a high affinity for interleukin-5 (IL-5), the key cytokine responsible for the maturation, recruitment, and survival of eosinophils. Unlike its predecessors, depemokimab has been engineered with specific modifications to its Fc region to extend its half-life significantly. This distinctive molecular structure allows the drug to maintain therapeutic concentrations over a six-month period.

By neutralizing IL-5, depemokimab inhibits the eosinophilic inflammatory cascade at its source. For patients with a documented history of eosinophilia, this targeted approach minimizes airway remodeling, reduces mucus hypersecretion, and decreases the bronchial hyperresponsiveness that leads to clinical exacerbations.

The SWIFT Phase III Program: Clinical Evidence and Efficacy

The FDA approval was primarily supported by data from the SWIFT-1 and SWIFT-2 clinical trials. These identical Phase III, randomized, double-blind, placebo-controlled studies evaluated the efficacy and safety of depemokimab in patients with severe asthma and a phenotype consistent with type 2 inflammation.

Study Design and Population

The trials enrolled individuals aged 12 years and older who were receiving medium- or high-dose inhaled glucocorticoids (ICS) plus at least one additional controller medication. Participants were required to have a history of at least two asthma exacerbations in the previous year or a blood eosinophil count of ≥300 cells/µL in the previous 12 months (or ≥150 cells/µL at screening). Patients were randomized to receive either depemokimab 100 mg subcutaneously every 26 weeks or a matching placebo, as add-on therapy to their existing standard of care.

Primary and Secondary Outcomes

The primary endpoint for both studies was the annualized rate of clinically significant asthma exacerbations over a 52-week period. The results were robust and statistically significant:

  • In SWIFT-1, depemokimab reduced the rate of asthma exacerbations by 58% compared to placebo (P < .001).
  • In SWIFT-2, a 48% reduction in exacerbations was observed (P < .001).
  • The annualized exacerbation rates were 0.46 for depemokimab vs. 1.11 for placebo in SWIFT-1, and 0.56 vs. 1.08 in SWIFT-2.

Beyond the primary endpoint, the trials demonstrated significant improvements in secondary metrics. Notably, there was a substantial reduction in exacerbations requiring emergency department (ED) visits or hospitalizations. This finding is particularly relevant for health policy experts and clinicians, as it translates to a direct reduction in the most acute and costly aspects of asthma care.

Safety and Tolerability Profile

Throughout the 52-week study period, depemokimab demonstrated a safety profile comparable to placebo. The most frequently reported adverse events (≥1% and more common than placebo) included:

  • Upper respiratory tract infections (e.g., common cold, sinusitis)
  • Headache
  • Injection-site reactions (e.g., pain, erythema, swelling)
  • Pruritus (itching)
  • Influenza-like symptoms

There were no significant differences in the incidence of serious adverse events between the treatment and control groups. The low frequency of injection-site reactions is particularly notable given the concentrated 100 mg dose required for six-month efficacy.

Expert Commentary: Clinical Utility and Adherence

The introduction of a twice-yearly biologic addresses one of the most persistent hurdles in chronic disease management: treatment adherence. In severe asthma, missing a dose of a biologic can lead to a rebound in eosinophil levels and a subsequent increase in the risk of life-threatening attacks. By aligning the treatment schedule with biannual clinical visits, depemokimab simplifies the therapeutic regimen for both patients and providers.

Clinicians should note that depemokimab is specifically indicated for the eosinophilic phenotype. Correct patient selection—utilizing blood eosinophil counts and exacerbation history—is essential to ensuring optimal outcomes. While the drug is highly effective for T2-high asthma, it is not indicated for the treatment of acute bronchospasm or status asthmaticus.

Conclusion: A Shift in the Management Paradigm

The FDA approval of depemokimab (Exdensur) represents a landmark achievement in respiratory medicine. By combining potent IL-5 inhibition with a convenient twice-yearly dosing schedule, it provides a unique solution for patients with severe eosinophilic asthma who have previously struggled with disease control or frequent administration schedules.

As the first long-acting biologic in this space, depemokimab not only reduces the clinical burden of exacerbations but also offers the potential for improved long-term disease stability. Future research will likely focus on the long-term impact of sustained eosinophil suppression on airway remodeling and the potential for disease modification in younger populations.

References

1. GlaxoSmithKline. (2024). FDA Approves Exdensur (depemokimab-ulaa) for Severe Asthma. Press Release.
2. SWIFT-1 and SWIFT-2 Investigators. Phase III Trials of Depemokimab in Severe Eosinophilic Asthma. (ClinicalTrials.gov Identifiers: NCT04719832, NCT04718103).
3. Pavord, I. D., et al. (2023). The Role of IL-5 in Severe Asthma: From Pathophysiology to Precision Medicine. The Lancet Respiratory Medicine.
4. FDA Prescribing Information: EXDENSUR (depemokimab-ulaa) injection, for subcutaneous use.
5. Full prescribing information is available here:https://gskpro.com/content/dam/global/hcpportal/en_US/Prescribing_Information/Exdensur/pdf/EXDENSUR-PI-PIL.PDF

Comments

No comments yet. Why don’t you start the discussion?

Leave a Reply