Biotech
152923-56-3
Relapsing Multiple Sclerosis (RMS)
Daclizumab represents a compelling and cautionary chapter in the history of biopharmaceutical development. A humanized monoclonal antibody, it was engineered with a novel and highly specific mechanism of action targeting the interleukin-2 (IL-2) receptor pathway. Its trajectory spanned two decades and two distinct therapeutic indications, beginning with a successful tenure in transplant medicine as Zenapax and culminating in its reinvention as Zinbryta, a potent, once-monthly therapy for relapsing forms of multiple sclerosis (MS).[1]
The promise of Daclizumab for MS was underpinned by robust clinical trial data. The pivotal Phase IIb SELECT and Phase III DECIDE trials demonstrated remarkable efficacy, significantly reducing relapse rates, disability progression, and radiological disease activity. Notably, in the DECIDE trial, Daclizumab proved superior not only to placebo but also to a first-line active comparator, interferon beta-1a, establishing it as a powerful new option for patients with inadequate responses to other treatments.[1] This efficacy was directly linked to its unique immunomodulatory effect: a re-balancing of the immune system that involved both the suppression of pathogenic T-cells and the expansion of regulatory Natural Killer (NK) cells.[6]
However, this profound efficacy was inextricably linked to a catastrophic risk profile that emerged over time. Initial concerns centered on severe hepatotoxicity, including cases of autoimmune hepatitis and liver failure, which prompted a prominent Boxed Warning from the U.S. Food and Drug Administration (FDA) and the implementation of a stringent Risk Evaluation and Mitigation Strategy (REMS) program.[2] The final, decisive blow came after its launch, with post-marketing reports of rare but severe and often fatal inflammatory brain disorders, including encephalitis and meningoencephalitis.[11] These events revealed that the very mechanism that so effectively controlled MS in the central nervous system could also trigger devastating secondary autoimmune attacks on other tissues.
Faced with an evolving and unacceptable benefit-risk profile, the manufacturers Biogen and AbbVie voluntarily withdrew Daclizumab from the global market in March 2018.[15] The story of Daclizumab thus serves as a critical case study in modern drug development. It illustrates the profound challenge of balancing the therapeutic potential of novel immunotherapies against rare but severe risks that may only become fully apparent after widespread clinical use. It underscores the limitations of pre-market safety assessments for detecting low-frequency events and affirms the paramount importance of rigorous, continuous pharmacovigilance in protecting public health.
Daclizumab is a biotechnology-derived drug, classified as a whole, humanized monoclonal antibody of the immunoglobulin G1 (IgG1) subtype.[1] It was developed by PDL BioPharma (then Protein Design Labs) through the humanization of a murine (mouse) monoclonal antibody known as anti-Tac.[1] This process was essential to minimize the potential for immunogenic reactions in human recipients. The final structure is a chimaera, composed of approximately 90% human and 10% murine protein sequences.[6] The human components were derived from the constant domains of human IgG1 and the variable framework regions of the human Eu myeloma antibody. The smaller murine portion consists of the complementarity-determining regions (CDRs)—the specific parts of the antibody that bind to the target—which were taken from the original anti-Tac antibody.[6]
Structurally, Daclizumab consists of two identical humanized gamma-1 heavy chains and two identical humanized kappa light chains, with a total molecular weight of approximately 144 kilodaltons (kDa).[6] It is produced using recombinant DNA technology in a mammalian cell line (NS0) within an animal component-free medium.[6]
For its use in multiple sclerosis under the trade name Zinbryta, Daclizumab was supplied as a sterile, preservative-free, colorless to slightly yellow, and clear to slightly opalescent solution for injection. It was available in a single-dose prefilled autoinjector or a single-dose prefilled syringe, each delivering 150 mg of the drug in a 1 mL volume.[6]
Table 1: Key Properties of Daclizumab (DB00111)
Characteristic | Value/Description | Source(s) |
---|---|---|
Drug Name | Daclizumab | 1 |
DrugBank ID | DB00111 | 1 |
Type | Biotech, Whole Antibody | 1 |
CAS Number | 152923-56-3 | 1 |
Molecular Weight | Approx. 144 kDa | 6 |
Source | Humanized (from murine mAb anti-Tac) | 1 |
Antibody Subtype | Humanized IgG1 | 6 |
Target | IL-2 Receptor Alpha Subunit (CD25) | 1 |
Formulation | 150 mg/mL solution for injection | 6 |
Trade Names | Zinbryta (MS), Zenapax (transplant, discontinued) | 1 |
The pharmacokinetic profile of Daclizumab was characterized by slow absorption, limited distribution, and a long elimination half-life, consistent with a large therapeutic protein.
Absorption: Following a single 150 mg subcutaneous (SC) injection, Daclizumab is slowly absorbed, reaching maximum serum concentrations (Tmax) in approximately 5 to 7 days.[1] A cross-study analysis determined its absolute bioavailability to be high, at about 90%.[1]
Distribution: When administered as a once-monthly 150 mg SC injection, Daclizumab accumulates to a level approximately 2.5 times that of a single dose, achieving steady-state serum concentrations by the fourth dose (i.e., after approximately four months of treatment).[6] At steady state, the mean maximum concentration (
Cmax) is approximately 30 µg/mL, and the mean minimum concentration (Cmin) is approximately 15 µg/mL.[6] The steady-state volume of distribution (
Vd) is approximately 6.34 L, indicating that the drug's distribution is largely confined to the vascular and interstitial fluid compartments, with limited penetration into deep tissues.[6]
Metabolism: As a large protein-based therapeutic, Daclizumab is not metabolized by the cytochrome P450 (CYP450) enzyme system in the liver.[1] Instead, it is presumed to be degraded through general protein catabolism, where proteases break it down into smaller peptides and constituent amino acids, which are then recycled by the body. This is the same pathway through which endogenous immunoglobulins are cleared.[1]
Elimination: Daclizumab exhibits slow clearance from the body, with a population pharmacokinetic analysis estimating the clearance rate at 0.212 L/day.[6] This slow clearance results in a long terminal elimination half-life of approximately 21 days, with a reported range of 11 to 38 days.[1] This long half-life provided the rationale for the convenient once-monthly dosing schedule. However, it also meant that the biological effects of the drug, and therefore the risk of adverse events, could persist for 8 to 12 weeks after the last dose was administered.[12] In patients who developed neutralizing antibodies against the drug, clearance was observed to be, on average, 19% higher.[6]
Table 2: Summary of Pharmacokinetic Parameters
Parameter | Value | Source(s) |
---|---|---|
Bioavailability (SC) | ~90% | 1 |
Time to Peak Concentration (Tmax) | 5-7 days | 1 |
Elimination Half-life (t1/2) | ~21 days (range 11-38 days) | 1 |
Volume of Distribution (Vd) | ~6.34 L | 6 |
Clearance (CL) | ~0.212 L/day | 6 |
Time to Steady State | By the 4th monthly dose | 1 |
The therapeutic efficacy and the eventual downfall of Daclizumab are both rooted in its unique and powerful modulation of the interleukin-2 (IL-2) signaling pathway. Its mechanism was not one of simple immunosuppression but rather a profound and specific re-balancing of distinct immune cell populations, which ultimately proved to be a double-edged sword.
The IL-2 system is a cornerstone of the adaptive immune response, acting as a master regulator of lymphocyte activation, proliferation, survival, and differentiation.[2] The cellular response to IL-2 is dictated by the composition of the IL-2 receptor (IL-2R) on the cell surface. This receptor is assembled from up to three distinct protein chains:
These chains combine to form three functionally distinct receptors:
Daclizumab was designed to bind with high affinity to the CD25 subunit (also known as the Tac subunit) of the IL-2R.[1] By occupying this site, it physically prevents IL-2 from binding to and signaling through the high-affinity receptor complex.[7] This targeted blockade has several direct and profound consequences on the immune system:
The most innovative and ultimately pivotal aspect of Daclizumab's mechanism was an indirect effect of its primary action. Activated T-cells and Tregs are major consumers of IL-2 in the body. By blocking their ability to bind IL-2 via the high-affinity receptor, Daclizumab dramatically reduces the overall consumption of this cytokine.[20] This leads to a significant increase in the bioavailability of free IL-2 in the serum.[20]
This surplus IL-2 becomes available to signal through the intermediate-affinity IL-2R (CD122/CD132), which is not blocked by Daclizumab as it lacks the CD25 subunit.[1] A key cell population that expresses this intermediate-affinity receptor is the CD56bright subset of NK cells. These cells have immunoregulatory properties and are distinct from the more cytotoxic CD56dim NK cells. The increased IL-2 signal drives a dramatic expansion and activation of these CD56bright NK cells.[1] In fact, the therapeutic efficacy observed in MS clinical trials was found to directly correlate with the degree of this NK cell expansion.[23] These activated, immunoregulatory NK cells are believed to contribute to the therapeutic effect by gaining access to the intrathecal compartment and selectively eliminating autologous activated T-cells.[6]
This complex interplay reveals that Daclizumab's mechanism was far more nuanced than simple immunosuppression. It did not broadly weaken the immune system; rather, it initiated a powerful immunomodulatory shift, actively suppressing pathogenic T-cell responses while simultaneously amplifying a regulatory NK cell response. This re-balancing was the source of its impressive clinical efficacy. However, this same forceful manipulation of the immune system is also what likely created the conditions for the severe, organ-specific autoimmune toxicities that led to its withdrawal. By concurrently inhibiting Tregs—the immune system's primary "peacekeepers" responsible for maintaining self-tolerance—and activating cytotoxic NK cells, Daclizumab created a state of profound immune dysregulation. In susceptible individuals, this dysregulated state could manifest as a loss of tolerance and a new autoimmune attack on self-antigens in organs such as the liver and brain, explaining the emergence of autoimmune hepatitis and inflammatory encephalitis.[8] The drug's benefit and its risk were, therefore, two outcomes of the exact same powerful mechanism.
The approval of Daclizumab for relapsing forms of multiple sclerosis was based on a robust clinical development program that demonstrated significant and superior efficacy in reducing disease activity. The key evidence came from two pivotal trials, SELECT and DECIDE, supported by long-term extension studies.
The SELECT trial was a 52-week, randomized, double-blind, placebo-controlled study designed to establish proof-of-concept and evaluate the efficacy and safety of Daclizumab High-Yield Process (HYP) in 600 patients with relapsing-remitting MS (RRMS).[4] Patients were randomized to receive subcutaneous Daclizumab HYP 150 mg, Daclizumab HYP 300 mg, or placebo once every 4 weeks.
The trial met its primary and key secondary endpoints, demonstrating a powerful effect on clinical and radiological measures of MS activity:
Table 3: Key Efficacy Endpoints from the SELECT Trial (vs. Placebo at 52 Weeks)
Endpoint | Daclizumab 150mg | Daclizumab 300mg | Placebo | p-value (vs. Placebo) | Source(s) |
---|---|---|---|---|---|
Annualized Relapse Rate (ARR) | 0.21 | 0.23 | 0.46 | <0.001 | 4 |
Proportion Relapse-Free | 81% | 80% | 64% | <0.001 | 4 |
Risk Reduction in 3-Month Sustained Disability Progression | 57% | 43% | N/A | p=0.021 | 4 |
Reduction in New/Newly Enlarging T2 Lesions | 70% | 79% | N/A | <0.001 | 27 |
Reduction in New Gd+ Lesions | 69% | 78% | N/A | <0.001 | 27 |
Following the success of SELECT, the DECIDE trial was launched. It was the largest and longest head-to-head study in MS at the time, designed to demonstrate the superiority of Daclizumab over an established first-line therapy.[3] This two- to three-year, randomized, double-blind study compared the efficacy and safety of Daclizumab HYP 150 mg SC monthly (n=919) against intramuscular interferon beta-1a (Avonex) 30 mcg weekly (n=922) in patients with RRMS.[3]
The results of DECIDE were pivotal, establishing Daclizumab's superior efficacy:
Patients who completed the pivotal trials were eligible to enroll in long-term open-label extension studies, such as SELECTION (NCT00870740) and EXTEND (NCT01797965), which provided data for up to eight years of continuous treatment.[5] These studies confirmed that the robust clinical and radiological efficacy of Daclizumab was sustained over the long term.[5] However, this extended observation period was also critical in revealing the cumulative and evolving safety risks, including a fatal case of autoimmune hepatitis in the SELECTION trial, which foreshadowed the severe adverse events that would ultimately lead to the drug's withdrawal.[28]
Table 4: Key Efficacy Endpoints from the DECIDE Trial (Daclizumab vs. Interferon Beta-1a)
Endpoint | Daclizumab 150mg | Interferon beta-1a | Relative Reduction / Odds Ratio | p-value | Source(s) |
---|---|---|---|---|---|
Reduction in ARR | N/A | N/A | 45% reduction | <0.001 | 5 |
Reduction in New/Newly Enlarging T2 Lesions (at 96 weeks) | N/A | N/A | 54% reduction | <0.001 | 5 |
Proportion Achieving NEDA (at 96 weeks) | 24.6% | 14.2% | OR: 2.059 | <0.0001 | 30 |
Despite its demonstrated efficacy, the clinical use of Daclizumab was ultimately terminated due to an unacceptable safety profile characterized by severe and unpredictable immune-mediated adverse reactions affecting multiple organ systems.
In clinical trials, the most common adverse reactions reported more frequently with Daclizumab than with the active comparator (interferon beta-1a) included nasopharyngitis, upper respiratory tract infection, rash, influenza, dermatitis, eczema, oropharyngeal pain, bronchitis, and lymphadenopathy.[8]
The most significant safety concern identified during the clinical development program was the risk of severe, and potentially fatal, liver injury.[2]
The adverse event that ultimately sealed Daclizumab's fate emerged during post-marketing surveillance. In early 2018, a cluster of cases of severe inflammatory brain disorders was reported in patients treated with the drug, leading to its rapid withdrawal from the market.[11]
Table 5: Incidence of Key Adverse Events from Pivotal Clinical Trials
Adverse Event | Daclizumab 150mg (%) | Interferon beta-1a (%) | Placebo (%) | Source(s) |
---|---|---|---|---|
Serious Infections | 4% (DECIDE) | 2% (DECIDE) | 0% (SELECT) | 27 |
Serious Cutaneous Reactions | 2% (DECIDE) | <1% (DECIDE) | 0% (SELECT) | 27 |
ALT/AST Elevations >5x ULN | 6% (DECIDE) | 3% (DECIDE) | <1% (SELECT) | 27 |
Lymphadenopathy | 5% | N/A | N/A | 8 |
Depression/Depressed Mood | 10% | N/A | N/A | 10 |
Note: Data from different trials (DECIDE vs. interferon; SELECT vs. placebo) are presented for context. Direct comparison across trials is not intended.
The regulatory history of Daclizumab is unique, marked by two distinct market approvals for different indications nearly two decades apart, followed by a rapid and decisive global withdrawal driven by post-marketing safety signals.
First Life (Zenapax): Daclizumab first entered the market under the brand name Zenapax, manufactured by Hoffmann-La Roche. It received FDA approval in December 1997 for the prophylaxis of acute organ rejection in patients receiving kidney transplants, making it the first humanized monoclonal antibody to be approved anywhere in the world.[1] It was used as part of an immunosuppressive regimen alongside cyclosporine and corticosteroids.[1] In 2009, Zenapax was withdrawn from the market. This decision was based on commercial factors—namely, the availability of newer and better therapeutic options for transplant patients—and was not driven by safety or efficacy concerns at that time.[1]
Second Life (Zinbryta): The drug was repurposed for neurology, leading to its second approval for multiple sclerosis.
The period following its launch was marked by escalating safety concerns. The 2017 EMA review highlighted that unpredictable and potentially fatal immune-mediated liver injury could occur during treatment and for up to 6 months after discontinuation.[13]
The situation reached a critical point in early 2018 with the emergence of reports of fatal inflammatory brain disorders. On March 2, 2018, the manufacturers, Biogen and AbbVie, took the decisive step of announcing a voluntary worldwide withdrawal of Zinbryta from the market.[11] The companies stated that given the "nature and complexity of adverse events being reported, characterizing the evolving benefit/risk profile of ZINBRYTA will not be possible going forward given the limited number of patients being treated".[12]
Regulatory agencies acted swiftly. The EMA and FDA issued immediate safety alerts and initiated a recall, recommending that no new patients start the therapy and that all current patients be transitioned to alternative treatments in close consultation with their neurologists.[11] All ongoing clinical trials involving Daclizumab were also terminated.[13]
Table 6: Timeline of Key Regulatory and Safety Events for Daclizumab
Date | Event | Significance/Details | Source(s) |
---|---|---|---|
Dec 1997 | FDA approves Zenapax | First humanized mAb approved worldwide; for preventing kidney transplant rejection. | 1 |
2009 | Zenapax discontinued | Withdrawn from market due to commercial factors, not safety. | 1 |
May 27, 2016 | FDA approves Zinbryta | For relapsing MS, with a restricted indication, Boxed Warning for liver injury, and a mandatory REMS program. | 1 |
July 2016 | EMA approves Zinbryta | For relapsing forms of MS in the European Union. | 11 |
Oct 2017 | EMA restricts Zinbryta use | PRAC review of liver safety leads to restricted indication for patients who failed other therapies. | 11 |
Mar 2, 2018 | Voluntary Worldwide Withdrawal | Biogen and AbbVie announce withdrawal due to reports of serious inflammatory brain disorders. | 11 |
Mar 2018 | EMA/FDA issue safety alerts | Agencies recommend immediate suspension and recall; patients to be transitioned to other therapies. | 11 |
The safety profile of Daclizumab necessitated specific contraindications and a range of warnings regarding its use and potential interactions with other medications.
The use of Daclizumab was strictly contraindicated in certain patient populations due to unacceptable risks:
Beyond the Boxed Warning for hepatic injury, the product labeling included several other critical warnings:
The immunomodulatory nature of Daclizumab created the potential for significant interactions:
The story of Daclizumab is a powerful illustration of the promise and peril of developing highly targeted, novel immunotherapies. Its rise and fall offer critical lessons for clinical medicine, regulatory science, and the pharmaceutical industry, highlighting the intricate relationship between efficacy, mechanism, and safety.
The Efficacy-Toxicity Paradox: The central lesson from Daclizumab is that its profound therapeutic efficacy and its catastrophic toxicity were two sides of the same coin, both stemming directly from its unique mechanism of action. By specifically blocking the high-affinity IL-2 receptor, it achieved a potent and desirable immunomodulatory re-balancing: suppressing pathogenic T-cells while expanding regulatory NK cells. This mechanism delivered superior clinical and radiological outcomes compared to existing therapies. However, this same forceful manipulation—particularly the concurrent depletion of regulatory T-cells—disrupted the delicate state of immune tolerance. This created a permissive environment for the development of new, severe autoimmune attacks on other organs, manifesting as fatal hepatitis and encephalitis. The experience demonstrates that even highly specific and targeted interference with a central immune pathway can have unforeseen and devastating systemic consequences.
A Landmark Case for Pharmacovigilance: The Daclizumab saga is a textbook case for the indispensable role of post-marketing pharmacovigilance. The pre-market clinical trial program, which included over 2,200 patients, was sufficiently powered to detect the relatively common risk of liver injury.[10] However, it was statistically incapable of detecting the rare but fatal neuroinflammatory events, which occurred at a frequency of approximately 12 cases among the 8,000 patients treated worldwide post-approval.[11] This starkly illustrates that the true safety profile of a new drug, especially one with a novel mechanism, can only be fully characterized once it is used in a larger, more heterogeneous real-world population. The rapid detection of these events and the subsequent swift withdrawal of the drug underscore the life-saving importance of a robust and responsive global pharmacovigilance system.
Implications for MS Treatment and Drug Development: The withdrawal of such a highly effective agent left a significant therapeutic gap for patients with aggressive MS who had failed multiple other treatments. For the field of drug development, the Daclizumab experience has raised the bar for the safety evaluation of future immunomodulatory drugs. It highlights the urgent need for better preclinical models and predictive biomarkers that can identify individuals at high risk for developing severe immune-mediated adverse reactions. The focus must shift from merely assessing if a drug "works" to understanding the full spectrum of its immunological consequences.
The Lingering Scientific Value: Despite its ultimate clinical failure, the development and study of Daclizumab were not without value. The investigation into its mechanism provided invaluable and lasting insights into the complex immunology of multiple sclerosis. It particularly illuminated the previously underappreciated role of the innate immune system—specifically the CD56bright NK cell population—in regulating CNS autoimmunity.[7] While Daclizumab itself is no longer a therapeutic option, the knowledge gained from its journey continues to inform new research and has opened novel avenues for the development of future therapies that may be able to harness its benefits without repeating its tragic risks.
Published at: August 13, 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.