AZD7442 (Evusheld): A Comprehensive Monograph on a Long-Acting Monoclonal Antibody Combination for COVID-19
Executive Summary
AZD7442, commercially known as Evusheld, represents a landmark achievement in the global response to the COVID-19 pandemic. Developed by AstraZeneca, this biotech therapeutic is a combination of two long-acting monoclonal antibodies, tixagevimab and cilgavimab, designed to provide passive immunity against SARS-CoV-2. Its development was predicated on a critical unmet medical need: the protection of immunocompromised and other high-risk individuals who are unable to mount an adequate immune response to vaccination. Through sophisticated molecular engineering, including modifications to extend its half-life and enhance its safety profile, AZD7442 was positioned as the first long-acting antibody for pre-exposure prophylaxis (PrEP) against COVID-19.
The clinical development program for AZD7442 was robust, comprising three pivotal Phase III trials that evaluated its efficacy across the spectrum of disease prevention and treatment. The PROVENT trial unequivocally demonstrated its prophylactic efficacy, showing a 77% reduction in the risk of developing symptomatic COVID-19, with protection lasting at least six months. Conversely, the STORM CHASER trial failed to meet its primary endpoint for post-exposure prophylaxis, providing crucial evidence on the therapeutic window for passive immunization. The TACKLE trial successfully established its utility as an early outpatient treatment for mild-to-moderate COVID-19, significantly reducing the risk of progression to severe disease, particularly when administered within three days of symptom onset.
The safety profile of AZD7442 was generally favorable and well-tolerated, with injection site reactions being the most common adverse event. A numerical imbalance in serious cardiovascular adverse events was noted in clinical trials, leading to a specific warning for patients with pre-existing cardiovascular risk factors, although a causal relationship was never established.
Ultimately, the clinical utility of AZD7442 was defined by its interaction with the rapidly evolving SARS-CoV-2 virus. While it retained potent activity against ancestral strains and early variants of concern, including Delta, its efficacy was challenged by the emergence of the Omicron lineage. Although it maintained neutralizing activity against early Omicron subvariants like BA.2 and BA.5, its function was significantly diminished against the highly immune-evasive BQ and XBB subvariants that became dominant in late 2022 and early 2023. This loss of in vitro neutralization prompted the U.S. Food and Drug Administration (FDA) to withdraw its Emergency Use Authorization in January 2023.
The regulatory journey of Evusheld was marked by divergent global decisions. While the FDA acted decisively based on in vitro susceptibility data, the European Medicines Agency (EMA) has maintained its marketing authorisation, reflecting different regulatory philosophies and risk-benefit assessments. This monograph provides a comprehensive analysis of AZD7442, detailing its molecular design, mechanism of action, clinical evidence base, safety profile, and its complex regulatory history, framing it as a critical case study in the development of antibody therapeutics during a pandemic and highlighting the enduring need for such interventions for the world's most vulnerable populations.
I. Introduction: The Rationale and Design of a Long-Acting Antibody Prophylaxis
The emergence of the COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, spurred an unprecedented global effort to develop effective vaccines. While active immunization proved to be a cornerstone of the public health response, it soon became apparent that this strategy was insufficient for all segments of the population. A significant unmet medical need persisted for individuals who were unable to mount an adequate or protective immune response to vaccination, leaving them highly vulnerable to severe outcomes from COVID-19. This "vaccine-gap" created the clinical and public health imperative for an alternative prophylactic strategy: passive immunization through the administration of long-acting monoclonal antibodies.
The Unmet Need in Immunocompromised and High-Risk Populations
A substantial portion of the global population, estimated at approximately 2%, is considered at increased risk of an inadequate response to COVID-19 vaccines.[1] This vulnerable cohort includes individuals with moderate to severe immune compromise resulting from underlying medical conditions, such as hematologic malignancies or primary immunodeficiencies, or from the receipt of immunosuppressive medications for conditions like solid organ transplantation or autoimmune diseases.[3] Data from the United Kingdom, for example, suggested that approximately 500,000 people were immunocompromised, with studies like the OCTAVE trial indicating that nearly 40% of this group mount a low or undetectable immune response after two vaccine doses.[3] This population, therefore, required a protective modality that did not depend on their own immune system to generate a response.
The development of a long-acting antibody (LAAB) for pre-exposure prophylaxis (PrEP) was conceived as a direct solution to this challenge. By providing ready-made neutralizing antibodies, a LAAB could confer immediate, passive immunity, bypassing the need for the recipient's own B-cells and T-cells to respond to an antigen.[5] This approach represented a strategic shift in the pandemic response, moving beyond a universal vaccination strategy to a more targeted, precision-medicine approach tailored to the specific immunological needs of the most vulnerable.
AZD7442: Composition and Strategic Development
AZD7442, which was later given the commercial name Evusheld, was developed by AstraZeneca to fill this prophylactic gap.[9] It is a biotech product classified as a combination of two fully human, neutralizing IgG1κ monoclonal antibodies:
tixagevimab (AZD8895) and cilgavimab (AZD1061).[10]
The development pathway for AZD7442 exemplifies a successful and rapid academia-industry collaboration. The parent antibodies were discovered by scientists at Vanderbilt University Medical Center, who isolated them from the B-cells of convalescent patients who had recovered from natural SARS-CoV-2 infection.[7] This strategy leveraged the power of the natural human immune response to identify highly potent neutralizing antibodies. In June 2020, these antibodies were licensed to AstraZeneca for further optimization, clinical development, and commercialization.[7]
From its inception, AZD7442 was designed as a combination therapy. The decision to co-formulate two distinct antibodies was a prescient strategy aimed at mitigating the risk of viral escape and resistance. It was understood that single monoclonal antibody therapies are vulnerable to failure if a mutation arises in the specific epitope they target.[7] By combining two potent antibodies that bind to different, non-overlapping sites on the viral spike protein, AZD7442 was engineered to "evade potential resistance with the emergence of new SARS-CoV-2 variants".[4] This dual-targeting mechanism creates a higher genetic barrier to resistance, as the virus would need to acquire mutations in two separate locations simultaneously to escape neutralization by both antibodies. This design foresight was a scientifically sound approach that contributed to the drug's initial broad efficacy against multiple early variants of concern.
The final product, Evusheld, is co-packaged and administered as two separate, sequential intramuscular (IM) injections, one for each antibody component.[3]
Table 1: Key Characteristics of AZD7442 (Evusheld)
| Characteristic | Description | Source(s) |
|---|
| Drug Name (Code) | AZD7442 | 11 |
| Drug Name (Commercial) | Evusheld | 9 |
| DrugBank ID | DB15787 | [User Query] |
| Type | Biotech; Long-Acting Antibody (LAAB) Combination | 7 |
| Component 1 | Tixagevimab (AZD8895) | 10 |
| Component 2 | Cilgavimab (AZD1061) | 10 |
| Antibody Isotype | Human IgG1κ | 9 |
| Origin | Derived from B-cells of convalescent COVID-19 patients | 7 |
| Developer | AstraZeneca (licensed from Vanderbilt University Medical Center) | 7 |
II. Molecular Engineering, Mechanism of Action, and Pharmacokinetics
The clinical utility of AZD7442 is rooted in its sophisticated molecular design. AstraZeneca's optimization of the parent antibodies discovered at Vanderbilt University involved targeted genetic engineering to enhance their duration of action and improve their safety profile. These modifications, combined with the fundamental mechanism of viral neutralization, created a therapeutic agent uniquely suited for long-term prophylaxis.
Targeting the SARS-CoV-2 Spike Protein: A Dual-Antibody Approach
The primary mechanism of action of AZD7442 is the direct neutralization of the SARS-CoV-2 virus.[7] Both tixagevimab and cilgavimab are designed to bind to the spike (S) glycoprotein, the structure on the surface of the virus responsible for mediating entry into host cells.[20] Specifically, their target is the Receptor Binding Domain (RBD), a critical region within the S1 subunit of the spike protein.[9] The RBD is responsible for engaging with the human angiotensin-converting enzyme 2 (ACE2) receptor, the primary portal for viral entry into respiratory and other cells.[7]
By binding to the RBD, the antibodies physically obstruct the interaction between the spike protein and the ACE2 receptor, thereby blocking viral attachment and preventing the virus from infecting host cells.[7] This blockade of viral entry is the sole mechanism of its protective effect, as confirmed by cell culture studies showing little to no antibody-dependent cellular functions.[9]
A critical feature of the drug's design is that tixagevimab and cilgavimab bind to distinct, non-overlapping epitopes on the RBD.[12] This allows for simultaneous binding of both antibodies to the spike protein, which can result in synergistic neutralization and, as previously noted, raises the genetic barrier for the emergence of resistant viral variants.[7]
Engineering for Longevity and Safety: The YTE and TM Modifications
To transform the potent neutralizing antibodies into a viable long-acting prophylactic, AstraZeneca introduced two key sets of amino acid substitutions into the fragment crystallizable (Fc) region of the antibodies.[7] This rational antibody design was a balancing act, simultaneously enhancing a desired property (longevity) while suppressing a potentially detrimental one (unwanted immune activation).
Half-Life Extension (YTE Modification)
To achieve a long duration of action, a triple amino acid substitution known as the "YTE" modification (M257Y/S259T/T261E) was engineered into the Fc region of both tixagevimab and cilgavimab.[12] This modification significantly increases the binding affinity of the antibody to the neonatal Fc receptor (FcRn).[13] The FcRn plays a crucial role in the pharmacokinetics of IgG antibodies by acting as a salvage receptor. After being taken up into cells via non-specific pinocytosis, IgG antibodies bind to FcRn within the acidic environment of the endosome. This binding protects the antibody from being targeted for lysosomal degradation and instead recycles it back to the cell surface, where it is released into circulation at physiological pH.[23] By enhancing the affinity for FcRn, the YTE modification makes this recycling process more efficient, thereby dramatically extending the antibody's circulating half-life. This engineering feat is what transforms a standard monoclonal antibody into a "long-acting" one, with a durability of action more than triple that of conventional antibodies.[10]
Safety Enhancement (TM Modification)
While the Fc region is critical for determining half-life, it also mediates effector functions by binding to Fcγ receptors (FcγR) on immune cells and to complement proteins like C1q.[25] These interactions can trigger inflammatory responses such as antibody-dependent cell-mediated cytotoxicity (ADCC). During the early phase of the pandemic, there was a theoretical concern about the potential for Antibody-Dependent Enhancement (ADE) of disease, a phenomenon where suboptimal antibodies could paradoxically worsen the infection.[14]
To mitigate this potential risk, AstraZeneca introduced a second set of modifications, known as the "TM" modification (L243F/L235E/P331S), which reduces the binding of the Fc region to FcγR and complement C1q.[10] This proactive safety measure effectively "silences" the effector functions of the antibodies, ensuring that their protective effect is derived solely from viral neutralization and not from potentially harmful inflammatory pathways. This rational design choice demonstrates a sophisticated approach that optimized not only for efficacy and pharmacokinetics but also for potential immunopathology.
Pharmacokinetic Profile: Absorption, Distribution, and Extended Half-Life
AZD7442 is formulated for intramuscular (IM) administration.[3] As large biologic molecules (approximately 150 kDa), monoclonal antibodies are absorbed slowly from the injection site, primarily via the lymphatic system rather than direct entry into blood capillaries.[28] This results in a time to maximum plasma concentration that typically ranges from two to eight days, with a bioavailability between 50% and 80%.[28]
The YTE modification confers a markedly extended elimination half-life. Pharmacokinetic data for cilgavimab demonstrated a half-life of days.[9] Across multiple sources, the half-life for the combination is consistently reported to be approximately
85 to 90 days.[9] This durable pharmacokinetic profile is the foundation of its clinical use as a long-acting prophylactic, with data from the PROVENT trial demonstrating clinically meaningful protection for at least six months following a single administration.[3] Early projections based on its pharmacokinetics suggested protection could potentially last for up to 12 months.[7]
Pharmacokinetic modeling from the PROVENT and STORM CHASER trials confirmed that the observed serum concentration-time profiles over a 457-day period were consistent with this extended half-life.[29] The apparent volume of distribution for cilgavimab is 8.7 L, indicating that it is primarily confined to the vasculature and interstitial space, which is typical for large protein therapeutics.[9] Elimination occurs primarily through intracellular catabolism into amino acids, and as such, the drug is not subject to renal excretion, and its pharmacokinetics are not expected to be impacted by renal impairment.[9]
Despite the proven extended half-life, the clinical development program did not prospectively establish an optimal dose-response relationship for repeat dosing. The initial prophylaxis trials used a 300 mg dose, while the treatment trial used 600 mg.[10] As new, less susceptible variants emerged, regulatory bodies like the FDA and TGA recommended increasing the prophylactic dose to 600 mg and, in Australia's case, adding an option for repeat dosing every six months.[34] However, this was often based on pharmacokinetic modeling and in vitro data rather than prospective clinical trial evidence. The European product information explicitly notes that "no safety and efficacy data available on repeat dosing".[36] This knowledge gap contributed to regulatory uncertainty and divergent global policies, with the UK government citing "insufficient data on the duration of protection" against Omicron as a key reason for its decision not to procure the drug.[37] The rapid pace of the pandemic outstripped the ability of traditional clinical trials to formally validate long-term dosing strategies, leaving a critical disconnect between the drug's known pharmacokinetics and the clinical evidence base for maintaining protection over time.
III. The Clinical Development Program: A Tripartite Evaluation of Efficacy
The clinical efficacy of AZD7442 was rigorously evaluated in a comprehensive Phase III program comprising three pivotal, randomized, controlled trials. Each trial was designed to assess the drug's utility in a distinct clinical scenario: pre-exposure prophylaxis (PROVENT), post-exposure prophylaxis (STORM CHASER), and early outpatient treatment (TACKLE). The collective results of these studies provided the foundational evidence for its regulatory submissions worldwide and defined its clinical role.
PROVENT (NCT04625725): Establishing Efficacy in Pre-Exposure Prophylaxis (PrEP)
The PROVENT trial was the cornerstone of the AZD7442 clinical program, designed to determine its efficacy as a preventative agent before exposure to the virus.
- Trial Design and Population: PROVENT was a Phase III, randomized, double-blind, placebo-controlled, multi-center trial that enrolled 5,197 adult participants across 87 sites in the US, UK, Spain, France, and Belgium.[17] Participants were randomized in a 2:1 ratio to receive a single 300 mg intramuscular dose of AZD7442 (150 mg of tixagevimab and 150 mg of cilgavimab; n=3,460) or a saline placebo (n=1,737).[17] The study population was specifically selected to represent those who would benefit most from passive immunization; over 75% of participants had co-morbidities, such as immunosuppressive disease, diabetes, or severe cardiac disease, that are associated with an increased risk for severe COVID-19 or a potentially reduced response to vaccination.[17]
- Efficacy Outcomes: The trial successfully met its primary endpoint. In the primary analysis, AZD7442 demonstrated a statistically significant 77% relative risk reduction (RRR) in the incidence of symptomatic, RT-PCR-confirmed COVID-19 compared to placebo (95% Confidence Interval [CI]: 46, 90; p<0.001).[3] This robust protection was shown to be durable over time. A subsequent analysis with a median follow-up of six months showed a consistent and slightly higher
83% RRR.[29] The final 15-month analysis confirmed this long-term benefit, with an RRR of 83.0% at day 183, which waned to 46.3% by day 366, indicating persistent, though decreasing, protection over a full year.[29]
- Protection Against Severe Disease: Perhaps the most critical finding was the profound protection against severe outcomes. At the time of the primary analysis, there were no cases of severe or critical COVID-19 and no COVID-19-related deaths in the group that received AZD7442. In contrast, the placebo arm recorded three cases of severe COVID-19, which included two deaths.[3] By the final analysis at day 366, AZD7442 had reduced the risk of severe or critical COVID-19 by 91.4% compared to placebo.[29]
- Significance: The unequivocal success of the PROVENT trial provided the definitive proof-of-concept for AZD7442 as a long-acting prophylactic agent. It was the first non-vaccine antibody combination to demonstrate prevention of COVID-19 in a clinical trial and formed the primary evidentiary basis for its initial Emergency Use Authorizations and conditional marketing approvals across the globe.[5]
STORM CHASER (NCT04625972): Investigating Post-Exposure Prophylaxis (PEP) and its Limitations
The STORM CHASER trial was designed to evaluate whether AZD7442 could prevent symptomatic disease in individuals who had already been recently exposed to an infected person.
- Trial Design and Population: This Phase III trial enrolled 1,121 unvaccinated adults who had a confirmed exposure to a person with SARS-CoV-2 within the preceding eight days.[29] Participants were randomized 2:1 to receive a single 300 mg IM dose of AZD7442 (n=749) or placebo (n=372).[29]
- Efficacy Outcomes: In stark contrast to PROVENT, the STORM CHASER trial failed to meet its primary endpoint. AZD7442 demonstrated only a 33% reduction in the risk of developing symptomatic COVID-19 compared to placebo, a result that was not statistically significant (95% CI: -25.9, 64.7; p=0.212).[29]
- Subgroup Analysis and Interpretation: The failure of the primary endpoint was revealing. A critical pre-planned subgroup analysis focused only on participants who were confirmed to be SARS-CoV-2 PCR-negative at the time of dosing (i.e., they were exposed but not yet infected). In this subgroup, AZD7442 showed a much more promising 73% RRR against symptomatic COVID-19.[16] This finding suggests that a significant number of participants in the overall trial may have already been in the earliest, asymptomatic stages of infection at the time of administration, a point at which the viral load may have been too established for the antibody infusion to reliably prevent the onset of symptoms.
- Significance: The divergent outcomes of PROVENT and STORM CHASER provide a crucial lesson in the temporal dynamics of passive immunotherapy. The success in PROVENT and in the PCR-negative subgroup of STORM CHASER highlights that the intervention is highly effective when protective antibody titers are established prior to viral seeding. The failure in the overall STORM CHASER population demonstrates that there is a narrow therapeutic window for prophylaxis; once viral replication has begun, even at a subclinical level, the efficacy of passively administered antibodies for preventing symptomatic disease is substantially diminished. This effectively ruled out AZD7442 as a reliable "morning-after" intervention and solidified its role as a true pre-exposure prophylactic.
TACKLE (NCT04625725): Demonstrating Therapeutic Benefit in Mild-to-Moderate COVID-19
The TACKLE trial shifted the focus from prevention to treatment, assessing the efficacy of AZD7442 in non-hospitalized patients with early, mild-to-moderate COVID-19.
- Trial Design and Population: TACKLE was a Phase III, randomized, double-blind, placebo-controlled trial that enrolled 903 non-hospitalized adults with mild-to-moderate COVID-19 who had been symptomatic for seven days or less.[10] A key feature of the trial was that 90% of participants had co-morbidities or were of an age that placed them at high risk of progressing to severe disease.[10] Participants received a single 600 mg IM dose of AZD7442 (300 mg of each antibody) or placebo.[10]
- Efficacy Outcomes: The trial successfully met its primary endpoint. A single dose of AZD7442 achieved a statistically significant 50% RRR in the risk of progressing to severe COVID-19 or death from any cause compared to placebo.[44]
- Impact of Early Intervention: The therapeutic benefit was highly dependent on the timing of administration. A pre-specified analysis revealed a clear gradient of efficacy:
- When administered within five days of symptom onset, AZD7442 reduced the risk of severe disease or death by 67%.[44]
- When administered within three days of symptom onset, the risk reduction was even greater, at 88%.[10]
- Significance: The positive results from TACKLE established a second major clinical indication for AZD7442: early outpatient treatment for high-risk individuals. This made Evusheld a unique therapeutic, as it became the only long-acting antibody combination to demonstrate Phase III efficacy in both prevention and treatment settings, a distinction that was particularly relevant for its approval in Europe.[10] The data also powerfully underscored a universal principle of antiviral therapy: early intervention is paramount. For a therapeutic like Evusheld to be maximally effective, it requires a healthcare ecosystem that facilitates rapid symptom recognition, immediate diagnostic testing, and prompt administration of the drug, a significant logistical consideration for its real-world implementation.
Table 2: Summary of Pivotal Phase III Clinical Trials for AZD7442
| Trial Name (Identifier) | Purpose | Key Population | N (AZD7442 / Placebo) | Primary Endpoint | Key Efficacy Result | Significance/Conclusion |
|---|
| PROVENT (NCT04625725) | Pre-Exposure Prophylaxis (PrEP) | High-risk adults (≥18 years) | 3,460 / 1,737 | Incidence of symptomatic COVID-19 | 77% RRR at primary analysis; 83% RRR at 6 months | Demonstrated high, durable efficacy for PrEP, forming the basis for global authorizations. |
| STORM CHASER (NCT04625972) | Post-Exposure Prophylaxis (PEP) | Unvaccinated adults recently exposed | 749 / 372 | Incidence of symptomatic COVID-19 | 33% RRR (Not statistically significant) | Failed to meet primary endpoint, suggesting limited utility for PEP and highlighting the importance of pre-exposure administration. |
| TACKLE | Treatment | Non-hospitalized, mild-moderate COVID-19 | 452 / 451 | Progression to severe COVID-19 or death | 50% RRR overall; 88% RRR if treated ≤3 days from symptom onset | Demonstrated efficacy as an early outpatient treatment, establishing a dual role for the drug. |
IV. Comprehensive Safety and Tolerability Profile
A thorough assessment of the safety and tolerability of a prophylactic agent is paramount, particularly when it is intended for long-term use in vulnerable populations. The clinical development program for AZD7442 provided a substantial body of safety data, which indicated that the drug was generally well tolerated, though it also revealed a specific cardiovascular safety signal that warranted careful consideration by regulatory agencies and clinicians.
Analysis of Common and Serious Adverse Events (AEs)
Across the pivotal Phase III trials, the overall safety profile of Evusheld was favorable and comparable to placebo.[10] The incidence of adverse events (AEs) was generally balanced between the treatment and control arms. In the PROVENT trial, AEs were reported in 35% of participants receiving Evusheld and 34% receiving placebo.[3] In the STORM CHASER trial, the rates were 46.5% and 51.9%, respectively.[29] Notably, in the TACKLE treatment trial, AEs were less frequent in the Evusheld group (29%) compared to the placebo group (36%).[10] This difference was largely attributable to the therapeutic effect of the drug; by reducing the severity of the illness, Evusheld decreased the incidence of COVID-19-related complications, such as COVID-19 pneumonia, which was reported in 6% of the Evusheld group versus 11% of the placebo group.[10] This finding powerfully illustrates that the drug's benefit extended to reducing overall morbidity, preventing more adverse events related to the disease than it caused.
- Most Common Adverse Reactions: The most frequently reported drug-related adverse reaction was injection site reaction, which included symptoms such as pain, erythema (redness), pruritus (itching), and induration (swelling) at the injection site. The incidence of these reactions was low, occurring in approximately 1.3% to 2.4% of participants who received Evusheld.[3] Other commonly reported adverse events, such as
headache, fatigue, and cough, occurred at rates similar to those in the placebo group, suggesting they were likely background events rather than drug-specific side effects.[4]
- Serious Adverse Events (SAEs): The overall rate of SAEs was low and did not show a concerning imbalance between the treatment and placebo arms. In the PROVENT trial, SAEs were reported in 6.2% of Evusheld recipients and 5.6% of placebo recipients, while in STORM CHASER, the rates were 2.7% and 4.3%, respectively.[29]
Special Warnings and Precautions
The product information for Evusheld includes several standard warnings and precautions consistent with its class as an injectable IgG1 monoclonal antibody.
- Hypersensitivity Including Anaphylaxis: Serious hypersensitivity reactions, including life-threatening anaphylaxis, are a known, albeit rare, risk with IgG1 monoclonal antibodies.[4] Consequently, Evusheld is contraindicated in individuals with a history of severe hypersensitivity to its active substances or excipients.[45] Administration is required to take place in a healthcare setting where severe reactions can be appropriately managed, and patients must be observed for at least one hour following injection.[4] A theoretical risk of cross-hypersensitivity with certain COVID-19 vaccines also exists, as Evusheld contains the excipient polysorbate 80, which is structurally similar to polyethylene glycol (PEG), an ingredient in some mRNA vaccines that has been associated with allergic reactions.[2]
- Clinically Significant Bleeding Disorders: As with any intramuscular injection, Evusheld should be administered with caution to individuals with thrombocytopenia (low platelet count) or any coagulation disorder, or to those taking anticoagulant medications, due to the risk of bleeding or hematoma at the injection site.[2]
- Use in Specific Populations:
- Pregnancy and Lactation: There are insufficient data from clinical trials to establish the safety of Evusheld during pregnancy or lactation. Its use in these populations is recommended only if the potential benefit to the mother is judged to outweigh the potential risk to the fetus or infant.[2]
- Pediatric Use: Evusheld is authorized for use in adolescents aged 12 years and older who weigh at least 40 kg. Its safety and effectiveness have not been established in younger children or those weighing less than 40 kg.[4]
In-depth Analysis of the Cardiovascular Safety Signal
The most significant safety finding to emerge from the clinical trial program was a numerical imbalance in serious cardiovascular adverse events.
- Trial Data: In the PROVENT trial, a higher rate of cardiac SAEs was observed in participants who received Evusheld (1.6%) compared to those who received placebo (0.9%).[36] These events included myocardial infarction (one of which was fatal) and cardiac failure.[45] A similar imbalance was seen for thromboembolic SAEs (0.9% in the Evusheld group vs. 0.4% in the placebo group).[45] A similar pattern was observed in the TACKLE trial, where four subjects in the Evusheld arm reported cardiac SAEs (including two MIs, one fatal) compared to one in the placebo arm.[46]
- Context and Causality: A crucial caveat, consistently highlighted by regulatory agencies, is that all of the subjects who experienced these events had pre-existing cardiovascular risk factors (such as hypertension or diabetes) and/or a prior history of cardiovascular disease.[4] The target population for Evusheld is inherently at a higher baseline risk for such events. Due to these confounding factors and the lack of a clear temporal pattern or plausible biological mechanism, a causal relationship between Evusheld and these cardiovascular events
has not been established.[4]
- Regulatory and Clinical Response: Despite the lack of a confirmed causal link, the consistency of this signal across trials was sufficient to prompt a specific warning in the product information. Regulators advised that clinicians should carefully consider the risks and benefits before initiating Evusheld in individuals at high risk for cardiovascular or thromboembolic events and that patients should be counseled to seek immediate medical attention if they experience suggestive symptoms.[4] This represents a classic regulatory approach to managing uncertainty: acknowledging a potential risk, alerting prescribers, and empowering them to make individualized risk-benefit assessments. This nuanced safety concern likely contributed to the cautious stance taken by some national health authorities when evaluating the drug for widespread procurement.[38]
Table 3: Summary of Adverse Events and Key Safety Warnings for Evusheld
| Part | Category | Details | Clinical Recommendation/Note |
|---|
| A: Adverse Reactions | Injection Site Reaction | Includes pain, erythema, pruritus, induration. Frequency: Common (≥1/100 to <1/10). | Generally mild to moderate and self-limiting. |
| Hypersensitivity | Includes rash, urticaria. Frequency: Common (≥1/100 to <1/10). | Monitor for signs of allergic reaction. |
| Injection Related Reaction | Includes headache, chills. Frequency: Uncommon (≥1/1,000 to <1/100). | Typically transient. |
| B: Key Warnings | Hypersensitivity/Anaphylaxis | Risk associated with IgG1 monoclonal antibodies. | Administer in a monitored setting; observe patient for at least 1 hour post-injection. Contraindicated in patients with a history of severe hypersensitivity. |
| Cardiovascular Events | Numerical imbalance of cardiac SAEs (MI, cardiac failure) and thromboembolic SAEs observed in trials. | Causal link not established. All affected patients had pre-existing risk factors. Weigh risk vs. benefit in individuals at high risk for cardiovascular events. |
| Bleeding Disorders | Standard precaution for IM injections. | Use with caution in patients with thrombocytopenia or those on anticoagulant therapy. |
V. The Shifting Landscape of Efficacy: Performance Against SARS-CoV-2 Variants
The clinical story of AZD7442 is inextricably linked to the evolutionary trajectory of its target, the SARS-CoV-2 virus. The drug's efficacy profile provides a real-time case study of the intense selective pressure a therapeutic agent can exert on a rapidly mutating RNA virus, demonstrating an evolutionary arms race that ultimately limited its clinical lifespan. Its journey from a broadly potent agent to one with limited utility against dominant strains highlights a critical challenge for the future of monoclonal antibody therapies in infectious diseases.
Initial Potency Against Ancestral Strains and Early Variants of Concern (VOCs)
The dual-antibody design of AZD7442, targeting two distinct and non-overlapping epitopes on the RBD, proved highly effective against the early forms of the virus. Preclinical and in vitro neutralization assays demonstrated potent activity against the ancestral USA-WA1/2020 strain of SARS-CoV-2.[13] More importantly, this design conferred resilience against the initial waves of viral evolution. AZD7442 retained potent neutralizing activity against the major early Variants of Concern (VOCs), including
Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), and Delta (B.1.617.2).[13] This broad initial efficacy was a key strength that supported its development and initial regulatory submissions, as the clinical trials were conducted during periods when these variants were predominant.
The Omicron Challenge: Diminished Neutralization and the Molecular Basis of Resistance
The emergence of the Omicron variant (B.1.1.529) in late 2021 represented a quantum leap in the evolution of the spike protein, which carried over 35 mutations compared to the ancestral strain.[21] This extensive antigenic drift posed the first significant challenge to AZD7442's efficacy.
- Differential Susceptibility Among Sublineages: Initial studies using live virus assays confirmed that AZD7442 retained neutralizing activity against the original Omicron variant, though at a reduced level compared to its activity against previous variants.[4] The inhibitory concentrations were still within the range observed in individuals who had recovered from natural infection, suggesting continued clinical relevance.[26] However, as the Omicron lineage diversified, a more complex picture emerged [12]:
- The BA.2 subvariant and its derivatives showed the highest susceptibility to AZD7442 among the early Omicron strains.
- The BA.1 and BA.1.1 subvariants showed lower susceptibility. This observation prompted the U.S. FDA in February 2022 to recommend doubling the initial prophylactic dose from 300 mg to 600 mg, with the rationale that a higher concentration of the antibodies might be necessary to overcome their reduced potency against these specific subvariants.[35]
- The BA.4 and BA.5 subvariants, which became dominant globally in mid-2022, exhibited an intermediate level of susceptibility, falling between that of BA.1 and BA.2.[12] The retained in vitro activity against BA.5 was a key piece of evidence supporting the expansion of Evusheld's approval for treatment in Europe during the BA.5 wave.[10]
- Molecular Mechanism of Reduced Susceptibility: The differential activity against Omicron sublineages was not random; it was traced to specific mutations within the antibody binding sites on the RBD. Molecular modeling and in vitro mutagenesis studies provided a clear structural explanation for the reduced susceptibility of the BA.1 lineage [12]:
- The G446S mutation, a signature of BA.1, was predicted to cause a steric clash with the binding site of cilgavimab.
- The Q493R mutation, present in BA.1 and most BA.2-derived subvariants, was predicted to cause a clash with the binding site of tixagevimab.
- The concurrent presence of both of these mutations in the BA.1 and BA.1.1 spike proteins was sufficient to explain their lower in vitro susceptibility to the AZD7442 combination.
Loss of Efficacy Against BQ and XBB Subvariants: The Tipping Point
While AZD7442 maintained a degree of activity against early Omicron subvariants, the continued and rapid evolution of the virus, leading to the emergence of sublineages derived from BA.2 and BA.5, ultimately rendered the drug ineffective.
- Emergence of Resistant Strains: In late 2022 and early 2023, new subvariants, including BQ.1, BQ.1.1, and the XBB lineage (including XBB.1.5), rose to dominance. These variants carried additional mutations on the RBD that further eroded the binding of monoclonal antibodies.
- Loss of Neutralization: Laboratory studies from multiple sources converged on a stark conclusion: AZD7442, along with other authorized monoclonal antibodies, was poorly effective or completely inactive against these newer strains. Data showed that the drug did not significantly neutralize BA.4.6, BQ.1, BQ.1.1, and XBB.[30] The highly transmissible and immune-evasive
XBB.1.5 subvariant, which became the dominant strain in the United States, was also not expected to be neutralized by Evusheld.[37]
- Clinical and Regulatory Impact: This comprehensive loss of in vitro neutralizing activity was the direct scientific trigger for the U.S. FDA's decision to withdraw the Emergency Use Authorization for Evusheld on January 26, 2023. The agency's rationale was that with over 90% of circulating infections in the U.S. being caused by resistant variants, the drug was no longer expected to provide any clinical benefit, and continuing its use would needlessly expose patients to potential side effects.[2]
The rapid decline in Evusheld's efficacy underscores a critical lesson for pandemic preparedness. Monoclonal antibody therapies, while powerful, are likely to have a finite lifespan against rapidly evolving pathogens. Their development and deployment must be tightly integrated with continuous, real-time molecular surveillance to track the emergence of resistance, and there must be a robust pipeline for the development of next-generation antibodies capable of neutralizing new variants. AstraZeneca's initiation of the SUPERNOVA trial for a next-generation LAAB is a direct response to this lesson.[2]
Table 4: Neutralization Activity of AZD7442 Against Key SARS-CoV-2 Variants
| SARS-CoV-2 Variant/Lineage | In Vitro Neutralizing Activity | Key Spike Mutations Impacting Binding | Clinical Implication / Regulatory Response |
|---|
| Ancestral (Wuhan, D614G) | Potent Neutralization | N/A | Basis for initial development and clinical trials. |
| Alpha, Beta, Gamma, Delta | Retained Potent Neutralization | N/A | Confirmed broad initial efficacy; supported use during early pandemic waves. |
| Omicron (BA.1 / BA.1.1) | Reduced Susceptibility | G446S (cilgavimab site), Q493R (tixagevimab site) | Prompted FDA recommendation to increase prophylactic dose to 600 mg. |
| Omicron (BA.2) | Retained Activity (Higher than BA.1) | Q493R | Supported continued use and confidence in the therapy. |
| Omicron (BA.4 / BA.5) | Intermediate Susceptibility | N/A | Supported continued use in Europe during the BA.5 wave. |
| Omicron (BQ.1, BQ.1.1, XBB, XBB.1.5) | No Significant Neutralization | Multiple additional RBD mutations | Led to the withdrawal of the FDA Emergency Use Authorization in the U.S. |
VI. A Global Regulatory Odyssey: Divergent Paths to Authorization and Withdrawal
The regulatory history of Evusheld is as complex and dynamic as the pandemic itself. As the scientific evidence evolved—from promising clinical trial results to concerning in vitro data on new variants—regulatory agencies in major global regions charted distinct and sometimes contradictory paths. This divergence in decision-making highlights fundamental differences in regulatory philosophy, risk tolerance, and the weighting of various forms of evidence, providing a compelling case study in the challenges of regulating therapeutics during a public health crisis.
United States (Food and Drug Administration - FDA)
The FDA's regulatory journey with Evusheld was characterized by rapid action based on emerging data, culminating in a decisive withdrawal of its authorization.
- Initial Authorization: On December 8, 2021, the FDA granted an Emergency Use Authorization (EUA) for Evusheld for pre-exposure prophylaxis (PrEP).[1] The authorization was for adults and adolescents (aged 12 and older, weighing at least 40 kg) with moderate-to-severe immune compromise or for whom vaccination was not recommended. This decision was based primarily on the robust efficacy and safety data from the PROVENT trial.[5]
- Dosage Update: As the Omicron BA.1 and BA.1.1 subvariants began to circulate, in vitro data suggested that a higher concentration of the antibodies might be needed to maintain efficacy. In response, on February 24, 2022, the FDA revised the EUA to increase the initial authorized dose from 300 mg (150 mg of each antibody) to 600 mg (300 mg of each antibody).[35]
- Authorization Withdrawal: The turning point came in late 2022 with the rise of the BQ and XBB subvariants. As in vitro data conclusively showed that Evusheld lacked neutralizing activity against these new strains, the FDA acted swiftly. On January 26, 2023, the agency announced that Evusheld was no longer authorized for emergency use in the U.S..[2] The rationale was precise and data-driven: with these resistant variants projected to be responsible for more than 90% of infections in the U.S., the drug was no longer expected to provide a clinical benefit. The agency concluded that continuing its use would expose patients to potential side effects without a reasonable expectation of efficacy.[37]
- Formal Revocation: The final chapter of the EUA was written based on a request from the manufacturer. On December 13, 2024, the FDA formally revoked the EUA for Evusheld.[60] This action was taken at the request of AstraZeneca, which informed the agency that all distributed lots of the product had expired and that the company no longer intended to offer Evusheld in the United States.[60]
European Union (European Medicines Agency - EMA)
The EMA's approach to Evusheld was more measured, resulting in a marketing authorisation that has remained active despite the challenges posed by new variants.
- Authorization for Prophylaxis: Following a rolling review that began in October 2021, the EMA's Committee for Medicinal Products for Human Use (CHMP) recommended approval, and a conditional marketing authorisation was granted on March 25, 2022, for the PrEP of COVID-19 in adults and adolescents.[18]
- Expansion to Treatment: In September 2022, the EMA expanded the authorisation to include the treatment of adults and adolescents with mild-to-moderate COVID-19 who are at increased risk of progressing to severe disease. This decision was based on the positive results of the TACKLE trial.[10]
- Status Amidst New Variants: Unlike the FDA, the EMA has not withdrawn the marketing authorisation for Evusheld.[2] The agency has acknowledged the laboratory studies showing that monoclonal antibodies, including Evusheld, are unlikely to be effective against emerging strains like BQ.1.1 and XBB.[56] However, instead of revoking the authorization, the EMA has issued guidance advising healthcare professionals to consider alternative treatments, especially if these resistant subvariants become prevalent in their region.[18] This approach reflects a different regulatory philosophy, one that prioritizes maintaining access to a therapeutic for a high-need population while empowering clinicians to make informed decisions based on local epidemiology and the totality of evidence.
United Kingdom (Medicines and Healthcare products Regulatory Agency - MHRA)
The UK's story is unique, involving a split between regulatory approval and a governmental decision not to procure the drug.
- Regulatory Approval: The MHRA granted a conditional marketing authorisation for Evusheld for PrEP on March 17, 2022.[3]
- Procurement Decision: Despite this regulatory green light, the UK government announced in August 2022 that it would not be procuring Evusheld for deployment through the National Health Service (NHS).[37] The government's rationale was multifaceted, citing "insufficient evidence" on the duration of protection against the then-dominant Omicron variants and "ongoing uncertainty about how these [in vitro tests] translate to clinical effectiveness".[38] This decision highlights how factors beyond regulatory approval, including national health policy, cost-effectiveness analyses, and interpretation of the evidence base, can influence a drug's availability.
Australia (Therapeutic Goods Administration - TGA)
Australia's TGA followed a path that included initial approval, subsequent expansion, and advisory updates in response to new variants.
- Initial Approval: The TGA granted provisional approval for Evusheld for PrEP in high-risk individuals on February 24, 2022.[22]
- Dosage and Indication Updates: On December 12, 2022, the TGA amended the provisional approval in two significant ways: it increased the recommended PrEP dosage to 600 mg with an option for repeat dosing every six months, and it expanded the approval to include the treatment of adults at increased risk of progressing to severe COVID-19.[34]
- Variant Advisory: Similar to the EMA, the TGA has not withdrawn the approval but has issued advisories. As of February 2023, the TGA noted that viral variants likely to be resistant to Evusheld were in widespread circulation in Australia and that this should be a key consideration for prescribers.[34] The regulatory status of the product, however, remains unchanged.[64]
Table 5: Timeline of Major Global Regulatory Actions for Evusheld
| Date | Regulatory Body (Region) | Action | Rationale / Key Data Source |
|---|
| Oct 2021 | EMA (EU) | Begins Rolling Review | Preliminary clinical study results 19 |
| Dec 8, 2021 | FDA (US) | Grants EUA for PrEP | PROVENT trial data 5 |
| Feb 24, 2022 | TGA (Australia) | Grants Provisional Approval for PrEP | PROVENT trial data 41 |
| Feb 24, 2022 | FDA (US) | Recommends Dose Increase to 600mg | In vitro data against Omicron BA.1/BA.1.1 35 |
| Mar 17, 2022 | MHRA (UK) | Grants Conditional Marketing Authorisation | PROVENT trial data 38 |
| Mar 25, 2022 | EMA (EU) | Grants Conditional Marketing Authorisation for PrEP | PROVENT trial data 61 |
| Aug 2022 | UK Government | Announces Decision Not to Procure | Insufficient efficacy data against Omicron variants 38 |
| Sep 2022 | EMA (EU) | Expands Authorisation to Treatment | TACKLE trial data 10 |
| Dec 12, 2022 | TGA (Australia) | Expands Approval to Treatment & Increases Dose | TACKLE trial data & emerging variant data 34 |
| Jan 26, 2023 | FDA (US) | Withdraws EUA | >90% of circulating variants (BQ, XBB) are resistant 37 |
| Dec 13, 2024 | FDA (US) | Formally Revokes EUA | Manufacturer request; all product expired 60 |
VII. Synthesis and Future Outlook
The development, deployment, and eventual limitation of AZD7442 (Evusheld) offers a profound and comprehensive case study on the lifecycle of a monoclonal antibody therapeutic in the crucible of a global pandemic. Its journey from a promising, rationally designed prophylactic to a therapy outpaced by viral evolution provides critical lessons for science, medicine, and public health policy. While its clinical utility against dominant SARS-CoV-2 strains has waned, the principles it established and the unmet need it sought to address remain as relevant as ever.
The Rise and Fall of AZD7442: Lessons in Monoclonal Antibody Development
AZD7442 was, by any scientific measure, a triumph of modern biotechnology. Its development was characterized by:
- Rational Design: The combination of two non-competing antibodies was a forward-thinking strategy to mitigate resistance.
- Sophisticated Engineering: The YTE and TM modifications successfully optimized the drug for longevity and safety, creating a true long-acting antibody.
- Rigorous Clinical Validation: The tripartite Phase III program (PROVENT, STORM CHASER, TACKLE) clearly defined its efficacy profile, demonstrating robust benefit in pre-exposure prophylaxis and early treatment.
Its "fall" was not due to a flaw in its design or a failure in its clinical performance against the viral strains it was tested against. Rather, its story is one of a successful drug that was ultimately overcome by the unprecedented speed and magnitude of antigenic drift in its target, SARS-CoV-2. The evolutionary leap represented by the Omicron variant and its subsequent rapid diversification into sublineages like BQ and XBB created a moving target that eventually escaped the neutralizing capacity of the two-antibody combination.
This experience yields several critical lessons for future pandemic preparedness. First, it validates the importance of targeting multiple, conserved epitopes to raise the barrier to resistance. Second, it underscores the absolute necessity of integrating therapeutic development with continuous, real-time genomic and virologic surveillance. The effectiveness of a monoclonal antibody is only as good as its activity against the currently circulating strains. Finally, it highlights the inherent challenge of conducting clinical trials at a pace that can keep up with viral evolution, which can lead to knowledge gaps regarding optimal dosing and scheduling in the face of new variants.
The Enduring Need for Passive Immunization Strategies
Despite the withdrawal of Evusheld's authorization in the United States, the fundamental clinical need for passive immunization strategies for immunocompromised and other vulnerable individuals has not diminished. On the contrary, the experience with Evusheld validated the entire concept. It provided definitive proof-of-concept that a long-acting antibody could safely and effectively protect this population when vaccines alone are insufficient.[29] The real-world data from its use provided a lifeline for many high-risk individuals, allowing them a degree of normalcy during the pandemic.[65] The problem that Evusheld was designed to solve—the "vaccine-gap"—persists, and the demand for a durable and effective prophylactic agent remains high.
The Next Generation: The SUPERNOVA Trial and the Future of LAABs
The story of AZD7442 does not end with its withdrawal but continues with the development of its successor. Acknowledging the lessons learned, AstraZeneca has initiated the SUPERNOVA Phase I/III trial (NCT05648110) to investigate a next-generation long-acting antibody.[2] This new therapeutic candidate has been specifically selected for its broader neutralizing capacity. Preclinical in vitro studies have shown that it can neutralize all SARS-CoV-2 variants tested to date, including the very strains that proved resistant to Evusheld.[2]
This iterative development cycle represents a new and necessary paradigm for creating therapeutics against rapidly evolving pathogens. It moves away from the model of developing a single "blockbuster" product and toward an adaptable platform-based approach. The foundational knowledge, manufacturing processes, and regulatory pathways established for Evusheld can now be leveraged to accelerate the development and review of its successors. The future of pandemic response will likely depend on such agile, iterative platforms that can be updated—much like annual influenza vaccines—to match the antigenic landscape of circulating pathogens, ensuring that the most vulnerable among us are never left without a shield.
Works cited
- EVUSHELD (formerly AZD7442) long-acting antibody combination authorized for emergency use in the US for pre-exposure prophylaxis (prevention) of COVID-19 - AstraZeneca US, accessed October 3, 2025, https://www.astrazeneca-us.com/media/press-releases/2021/evusheld-formerly-AZD7442-long-acting-antibody-combination-authorized-for-emergency-use-in-the-US-for-pre-exposure-prophylaxis-prevention-of-COVID-191.html
- Update on US Food and Drug Administration Emergency Use Authorization of EVUSHELD, accessed October 3, 2025, https://www.astrazeneca-us.com/media/press-releases/2023/update-on-us-food-and-drug-administration-emergency-use-authorization-of-evusheld.html
- AstraZeneca's antibody combination, Evusheld (tixagevimab co-packaged with cilgavimab) authorised for use in Great Britain for pre-exposure prophylaxis (prevention) of COVID-19, accessed October 3, 2025, https://www.astrazeneca.com/media-centre/press-releases/2022/astrazenecas-antibody-combination-evusheld-authorised-for-use-in-great-britain-for-pre-exposure-prophylaxis-prevention-of-covid-19.html
- EVUSHELD long-acting antibody combination retains neutralizing activity against Omicron variant in independent FDA study - AstraZeneca US, accessed October 3, 2025, https://www.astrazeneca-us.com/media/press-releases/2021/evusheld-long-acting-antibody-combination-retains-neutralizing-activity-against-omicron-variant-in-independent-fda-study.html
- EVUSHELD (formerly AZD7442) long-acting antibody combination authorized for emergency use in the US for pre-exposure prophylaxis (prevention) of COVID-19 | HFSA, accessed October 3, 2025, https://hfsa.org/evusheld-formerly-azd7442-long-acting-antibody-combination-authorized-emergency-use-us-pre-exposure
- Distribution and Patient Prioritization for Tixagevimab/Cilgavimab (Evusheld), accessed October 3, 2025, https://acphd-web-media.s3-us-west-2.amazonaws.com/media/health-alerts/evusheld-cdph-health-alert-2022.02.15.pdf
- COVID-19 Long-Acting AntiBody (LAAB) combination AZD7442 rapidly advances into Phase III clinical trials - NATAP, accessed October 3, 2025, https://natap.org/2020/COVID/010620_02.htm
- AZD7442 PROVENT Phase III Prophylaxis Trial Met Primary Endpoint in Preventing Symptomatic COVID-19 - CLL Society, accessed October 3, 2025, https://cllsociety.org/2021/08/azd7442-provent-phase-iii-prophylaxis-trial-met-primary-endpoint-in-preventing-symptomatic-covid-19/
- Cilgavimab: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 3, 2025, https://go.drugbank.com/drugs/DB16393
- Evusheld long-acting antibody combination approved in the EU for the treatment of COVID-19 - AstraZeneca, accessed October 3, 2025, https://www.astrazeneca.com/media-centre/press-releases/2022/evusheld-approved-in-eu-for-covid-19-treatment.html
- COVID-19 Dashboard | DrugBank Online, accessed October 3, 2025, https://go.drugbank.com/covid-19
- Molecular Characterization of AZD7442 (Tixagevimab-Cilgavimab) Neutralization of SARS-CoV-2 Omicron Subvariants - PMC, accessed October 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10100701/
- (PDF) The SARS-CoV-2 monoclonal antibody combination, AZD7442, is protective in non-human primates and has an extended half-life in humans - ResearchGate, accessed October 3, 2025, https://www.researchgate.net/publication/358110459_The_SARS-CoV-2_monoclonal_antibody_combination_AZD7442_is_protective_in_non-human_primates_and_has_an_extended_half-life_in_humans
- Update on US Food and Drug Administration Emergency Use Authorisation of Evusheld - AstraZeneca, accessed October 3, 2025, https://www.astrazeneca.com/media-centre/press-releases/2023/update-on-evusheld-us-eua.html
- FDA grants emergency use authorization to anti-SARS-COV-2 mAb Evusheld, accessed October 3, 2025, https://www.antibodysociety.org/covid-19/fda-grants-emergency-use-authorization-to-anti-sars-cov-2-mab-evusheld/
- AstraZeneca Covid-19 drug candidate fails in Phase III study - Clinical Trials Arena, accessed October 3, 2025, https://www.clinicaltrialsarena.com/news/astrazeneca-covid-drug-trial-data/
- PROVENT Trial: AZD7442 as Long-Acting Pre-Exposure Prophylaxis Protects Against Symptomatic COVID-19 - Contagion Live, accessed October 3, 2025, https://www.contagionlive.com/view/provent-trial-azd7442-pre-exposure-prophylaxis-symptomatic-covid-19
- Evusheld | European Medicines Agency (EMA), accessed October 3, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/evusheld
- EMA starts rolling review of Evusheld (tixagevimab and cilgavimab), accessed October 3, 2025, https://www.ema.europa.eu/en/news/ema-starts-rolling-review-evusheld-tixagevimab-cilgavimab
- Understanding the Molecular Actions of Spike Glycoprotein in SARS-CoV-2 and Issues of a Novel Therapeutic Strategy for the COVID-19 Vaccine - MDPI, accessed October 3, 2025, https://www.mdpi.com/2673-7426/4/2/84
- Molecular Characterization of AZD7442 (Tixagevimab-Cilgavimab) Neutralization of SARS-CoV-2 Omicron Subvariants - ResearchGate, accessed October 3, 2025, https://www.researchgate.net/publication/369037170_Molecular_Characterization_of_AZD7442_Tixagevimab-Cilgavimab_Neutralization_of_SARS-CoV-2_Omicron_Subvariants
- AstraZeneca's Evusheld™ (Tixagevimab and Cilgavimab) Granted Provisional Approval For Pre-Exposure Prophylaxis (Prevention) Of COVID-19 In Australia's Most At-Risk, accessed October 3, 2025, https://www.astrazeneca.com.au/news/media-releases/2022/astrazenecas-evusheld.html
- Pharmacokinetics of monoclonal antibodies and Fc-fusion proteins - Oxford Academic, accessed October 3, 2025, https://academic.oup.com/proteincell/article/9/1/15/6768236
- (PDF) Pharmacokinetics of Monoclonal Antibodies - ResearchGate, accessed October 3, 2025, https://www.researchgate.net/publication/317960099_Pharmacokinetics_of_Monoclonal_Antibodies
- Tutorial on Monoclonal Antibody Pharmacokinetics and Its Considerations in Early Development - PMC - PubMed Central, accessed October 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6226118/
- EVUSHELD long-acting antibody combination retains neutralizing activity against Omicron variant in studies from Oxford and Washington Universities - AstraZeneca US, accessed October 3, 2025, https://www.astrazeneca-us.com/media/press-releases/2021/evusheld-long-acting-antibody-combination-retains-neutralizing-activity-against-omicron-variant-in-studies-from-oxford-and-washington-universities.html
- Pharmacokinetics of Monoclonal Antibodies - PMC - PubMed Central, accessed October 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5613179/
- Monoclonal Antibodies: Clinical Pharmacology Keys | Applied Clinical Trials Online, accessed October 3, 2025, https://www.appliedclinicaltrialsonline.com/view/monoclonal-antibodies-clinical-pharmacology-keys
- Efficacy, Safety, and Pharmacokinetics of AZD7442 (Tixagevimab/Cilgavimab) for Prevention of Symptomatic COVID-19: 15-Month Final Analysis of the PROVENT and STORM CHASER Trials - PubMed, accessed October 3, 2025, https://pubmed.ncbi.nlm.nih.gov/38703336/
- Low neutralizing activity of AZD7442 against current SARS-CoV-2 Omicron variants in patients with B-cell malignancies - PMC, accessed October 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9906471/
- AZD7442 PROVENT Phase III prophylaxis trial met primary endpoint in preventing COVID-19 - AstraZeneca, accessed October 3, 2025, https://www.astrazeneca.com/media-centre/press-releases/2021/azd7442-prophylaxis-trial-met-primary-endpoint.html
- AZD7442 PROVENT Phase III prophylaxis trial met primary endpoint in preventing COVID-19 - AstraZeneca US, accessed October 3, 2025, https://www.astrazeneca-us.com/media/press-releases/2021/azd7442-provent-phase-iii-prophylaxis-trial-met-primary-endpoint-in-preventing-covid-19-08202021.html
- NEW MEXICO HEALTH ALERT NETWORK (HAN) ALERT Latest Guidance from FDA on AstraZeneca COVID-19 monoclonal antibody Evusheld DATE 1, accessed October 3, 2025, https://www.nmhealth.org/publication/view/general/7320/
- Use of Evusheld - Department of Health, Disability and Ageing, accessed October 3, 2025, https://www.health.gov.au/sites/default/files/2023-02/evusheld-fact-sheet-for-health-professionals.pdf
- FDA announces Evusheld is not currently authorized for emergency use in the U.S., accessed October 3, 2025, https://www.fda.gov/drugs/drug-safety-and-availability/fda-announces-evusheld-not-currently-authorized-emergency-use-us
- Evusheld, INN- tixagevimab, cilgavimab - European Medicines Agency, accessed October 3, 2025, https://www.ema.europa.eu/en/documents/product-information/evusheld-epar-product-information_en.pdf
- Covid-19: FDA removes US authorisation for antibody drug Evusheld | The BMJ, accessed October 3, 2025, https://www.bmj.com/content/380/bmj.p264
- Decision on Evusheld as a COVID-19 treatment - GOV.UK, accessed October 3, 2025, https://www.gov.uk/government/publications/decision-on-evusheld-as-a-coronavirus-covid-19-treatment-letter-to-patient-groups/decision-on-evusheld-as-a-covid-19-treatment
- AZD7442 Completed Phase 3 Trials for Coronavirus Disease 2019 (COVID‑19) Prevention, accessed October 3, 2025, https://go.drugbank.com/drugs/DB15787/clinical_trials?conditions=DBCOND0129755&phase=3&purpose=prevention&status=completed
- AstraZeneca announces Evusheld long-acting antibody combination approved in the EU for the pre-exposure prophylaxis (prevention) of COVID-19 in a broad population, accessed October 3, 2025, https://www.astrazeneca.com/country-sites/thailand/press-releases/astrazeneca-announces-evusheld-long-acting-antibody-combination-approved-in-the-eu-for-the-pre-exposure-prophylaxis-prevention-of-covid-19-in-a-broad-population.html
- TGA provisionally approves AstraZeneca's combination therapy ..., accessed October 3, 2025, https://www.tga.gov.au/news/media-releases/tga-provisionally-approves-astrazenecas-combination-therapy-tixagevimab-and-cilgavimab-evusheld-pre-exposure-prevention-prophylaxis-covid-19
- STORM CHASER Trial Fails to Meet Primary Endpoint in Post-Exposure Prevention of Symptomatic COVID-19 | American Pharmaceutical Review, accessed October 3, 2025, https://www.americanpharmaceuticalreview.com/1315-News/576828-STORM-CHASER-Trial-Fails-to-Meet-Primary-Endpoint-in-Post-Exposure-Prevention-of-Symptomatic-COVID-19/
- Evusheld significantly prevented COVID-19 disease progression or death in TACKLE Phase III treatment trial - AstraZeneca, accessed October 3, 2025, https://www.astrazeneca.com/country-sites/thailand/press-releases/evusheld-significantly-prevented-COVID-19.html
- AZD7442 reduced risk of developing severe COVID-19 or death in TACKLE Phase III outpatient treatment trial - AstraZeneca, accessed October 3, 2025, https://www.astrazeneca.com/media-centre/press-releases/2021/azd7442-phiii-trial-positive-in-covid-outpatients.html
- Product Information: Evusheld, accessed October 3, 2025, https://www.tga.gov.au/sites/default/files/evusheld-pi.pdf
- Summary of Product Characteristics for Evusheld - GOV.UK, accessed October 3, 2025, https://www.gov.uk/government/publications/regulatory-approval-of-evusheld-tixagevimabcilgavimab/summary-of-product-characteristics-for-evusheld
- Evusheld Injection: Uses, Dosage, Warnings, Side Effects - Drugs.com, accessed October 3, 2025, https://www.drugs.com/evusheld.html
- Patient Information Leaflet for Evusheld - GOV.UK, accessed October 3, 2025, https://www.gov.uk/government/publications/regulatory-approval-of-evusheld-tixagevimabcilgavimab/patient-information-leaflet-for-evusheld
- PRODUCT MONOGRAPH INCLUDING PATIENT MEDICATION INFORMATION EVUSHELDTM tixagevimab and cilgavimab injection solution, 100 mg/mL, accessed October 3, 2025, https://pdf.hres.ca/dpd_pm/00071063.PDF
- Molecular Characterization of AZD7442 (Tixagevimab-Cilgavimab) Neutralization of SARS-CoV-2 Omicron Subvariants - PubMed, accessed October 3, 2025, https://pubmed.ncbi.nlm.nih.gov/36877050/
- Update: Evusheld no longer authorized in the U.S. | Immune Deficiency Foundation, accessed October 3, 2025, https://primaryimmune.org/resources/news-articles/update-evusheld-no-longer-authorized-us
- Molecular Characterization of AZD7442 (Tixagevimab-Cilgavimab ..., accessed October 3, 2025, https://journals.asm.org/doi/10.1128/spectrum.00333-23
- TGA provisionally approves first prevention therapy for COVID-19 - RACGP, accessed October 3, 2025, https://www1.racgp.org.au/newsgp/clinical/tga-provisionally-approves-first-prevention-therap
- FDA Withdraws Emergency use Authorization of Evusheld - NPHIC, accessed October 3, 2025, https://www.nphic.org/news/news-highlights/1268-fda-withdraws-emergency-use-authorization-of-evusheld
- Ineffective Against Current Variants, Evusheld's FDA Authorization Revoked, accessed October 3, 2025, https://kffhealthnews.org/morning-breakout/ineffective-against-current-variants-evushelds-fda-authorization-revoked/
- ETF warns that monoclonal antibodies may not be effective against emerging strains of SARS-CoV-2 | European Medicines Agency (EMA), accessed October 3, 2025, https://www.ema.europa.eu/en/news/etf-warns-monoclonal-antibodies-may-not-be-effective-against-emerging-strains-sars-cov-2
- COVID-19 Update: Evusheld Unlikely to Neutralize XBB.1.5 Omicron Variant (online only), accessed October 3, 2025, https://secure.medicalletter.org/TML-article-1669e
- COVID-19 Prophylactic Evusheld Not Expected to Neutralize New Omicron XBB.1.5 Variant, accessed October 3, 2025, https://www.contagionlive.com/view/covid-19-prophylactic-evusheld-not-expected-to-neutralize-new-omicron-xbb-1-5-variant
- COVID-19 roundup: New omicron subvariants may be up to 21x more evasive against vaccine antibodies - Advisory Board, accessed October 3, 2025, https://www.advisory.com/daily-briefing/2023/01/20/covid-19-roundup
- December 13, 2024 AstraZeneca Pharmaceuticals LP ... - FDA, accessed October 3, 2025, https://www.fda.gov/media/184468/download
- EMA recommends authorisation of COVID-19 medicine Evusheld, accessed October 3, 2025, https://www.ema.europa.eu/en/news/ema-recommends-authorisation-covid-19-medicine-evusheld
- EMA recommends authorisation of COVID-19 medicine Evusheld - AIFA, accessed October 3, 2025, https://www.aifa.gov.it/en/-/ema-raccomanda-l-autorizzazione-di-evusheld-medicinale-anti-covid-19
- Australian public assessment report for Evusheld - Therapeutic Goods Administration (TGA), accessed October 3, 2025, https://www.tga.gov.au/sites/default/files/2023-01/auspar-evusheld-230104.pdf
- Update on effectiveness of monoclonal antibodies against COVID variants, accessed October 3, 2025, https://www.tga.gov.au/news/news/update-effectiveness-monoclonal-antibodies-against-covid-variants
- Implementation of AZD7442 (Tixagevimab/Cilgavimab) COVID-19 Pre-exposure Prophylaxis (PrEP) in the Largest Health Maintenance Organization in Israel: Real-world Uptake and Sociodemographic and Clinical Characteristics Across Immunocompromised Patient Groups, accessed October 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11128413/
- COVID Completed Phase 2 Trials for AZD7442 (DB15787) | DrugBank Online, accessed October 3, 2025, https://go.drugbank.com/indications/DBCOND0127821/clinical_trials/DB15787?phase=2&status=completed