Evolving Impact of Latest Monoclonal Antibodies as Pre-Exposure and Treatment in Immunocompromised and Elderly Patients with New Variants of Sars-Cov-2. A Review of Knows and Unknowns at December 2024 Download PDF

Journal Name : SunText Review of Virology

DOI : 10.51737/2766-5003.2024.059

Article Type : Research Article

Authors : Weimer LE, Cattari G, Fanales Belasio E, Cuccuru E and Vidili Gianpaolo

Keywords : SARS-CoV-2; Covid-19

Abstract

The most significant health challenge in the 21st century has been the Sars-Cov-2. The Extensive research and the Global Cooperation have provided a profound understanding of the latest therapies, fundamental biological and molecular characteristics of SARS-CoV-2. Pre-exposure prophylaxis using monoclonal antibodies is one complementary preventative therapy to reduce severity of breakthrough Sars-Cov-2 in persons with severe immunocompromise due organ transplant, cancer, HIV or use of certain medications experience diminished SARS-CoV-2 vaccine immune response and remain at higher risk for severe COVID-19 outcomes; across many studies, Monoclonal Antibodies Pre-Exposure is associated with a 60% to 80% reduction in severe COVID-19 outcomes. Covid-19 evolution, however, leads to viral mutations that can evade Monoclonal Antibodies due to the selective nature of their binding sites. Several Monoclonal Antibodies that received FDA emergency use authorization for prevention of Sars-Cov-2 have since had this authorization revoked once circulating variants demonstrated immune evasion. This Review explain the Next –Generation of Pre-Exposure Prophylaxis, the recommendations for the Clinical Management for Persons with Risk factors for COVID-19, include obesity, older age, underlying medical conditions such as diabetes, inadequate vaccination, immunocompromised condition


Background

The most significant health challenge in the 21st century has been the Sars-Cov-2. The Extensive research and the Global Cooperation have provided a profound understanding of the lastest therapies, fundamental biological and molecular characteristics of SARS-CoV-2 [1].

Pre-exposure prophylaxis using monoclonal antibodies is one complementary preventative therapy to reduce severity of breakthrough Sars-Cov-2 in persons with severe immunocompromise due organ transplant, cancer, HIV or use of certain medications experience diminished SARS-CoV-2 vaccine immune response and remain at higher risk for severe COVID-19 outcomes [2]; across many studies, Monoclonal Antobodies Pre-Exposure is associated with a 60% to 80% reduction in severe COVID-19 outcomes.

Covid-19 evolution, however, leads to viral mutations that can evade Monoclonal Antibodies due to the selective nature of their binding sites. Several Monoclonal Antibodies that received FDA emergency use authorization for prevention of Sars-Cov-2 have since had this authorization revoked once circulating variants demonstrated immune evasion [3]. This Review explain the Next –Generation of Pre-Exposure Prophylaxis, the recommendations for the Clinical Mamagement for Persons with  Risk factors for COVID-19 , include obesity, older age, underlying medical conditions such as diabetes, inadequate vaccination, immunocompromised condition.

Introduction

COVID-19 still represents a significant and disproportionate risk for immunocompromised patients, with infection often leading to serious and protracted illness. Vaccination against Sars-Cov-2 has reduced in the Word the burden Covid-19. The urgent need to find new therapeutic strategies to combat the Sars-Cov-2 pandemic led to the design of numerous anti-Covid 19 monoclonal antibody for Therapy and Pre-Exposure Prophylaxis [4].

The immunocompromised persons and those who cannot be vaccinated, remain at risk for severe Covid-19. As the world deals with this evolving threat, the narrative extends to the realm of emerging variants, each displaying new mutations with implications that remain largely misunderstood. Notably, the JN.1 Omicron lineage is gaining global prevalence, and early findings suggest it stands among the immune-evading variants, a characteristic attributed to its mutation L455S. Moreover, the detrimental consequences of the novel emergence of SARS-CoV-2 lineages bear a particularly critical impact on immunocompromised individuals and older adults. Immunocompromised individuals face challenges such as suboptimal responses to COVID-19 vaccines, rendering them more susceptible to severe disease. Similarly, older adults have an increased risk of severe disease and the presence of comorbid conditions, find themselves at a heightened vulnerability to develop COVID-19 disease. Thus, recognizing these intricate factors is crucial for effectively tailoring public health strategies to protect these vulnerable populations. In this context, this review aims to describe, analyze, and discuss the current progress of the next-generation treatments encompassing   the lastest monoclonal antibodies for Pre-Exposure Therapy immunotherapeutic approaches and advanced therapies emerging as complements that will offer solutions to counter the disadvantages of the existing options. Although Monoclonal Antibodies as Prophilaxis and Therapy report efficacy as between 50–85% [5-6], global access is currently largely inequitable.

Preliminary outcomes show that these strategies target the virus and address the immunomodulatory responses associated with COVID-19. Furthermore, the capacity to promote tissue repair has been demonstrated, which can be particularly noteworthy for immunocompromised individuals who stand as vulnerable actors in the global landscape of coronavirus infections [7-9].

Monoclonal antibodies, which protect against disease irrespective of immune system status and provide rapid protection, are potential options for Covid-19 immunoprophylaxis. Some combinations of monoclonal antibodies are already in use through emergency or temporary authorization for preexposure or postexposure prophylaxis against Covid-19 or treatment of mild-to-moderate disease. The emerging next-generation treatments possess broader potential, offering protection against a wide range of variants and enhancing the ability to counter the impact of the constant evolution of the virus. The main purpose of this leterature Review is to highlight the possible strategies to optimize and protect current and future therapeutic options Pre-Exposure to treat the most vulnerable patients [10-11].

Omicron Variant: "Almost a new Pandemic"

With the emergence of this variant, the researchers realized that vaccination offered less protection and that a third dose was needed to immunize the population. While previous variants had fewer than ten mutations on the spike protein compared with the original strain, Omicron had more than thirty. It was almost as if a new pandemic had begun. What the researchers were less aware of was that Omicron had a significant ability to evade monoclonal antibodies. This was demonstrated by several scientists in an article published in December 2021. In their research, the scientists showed that two thirds of the monoclonal antibodies used in clinical practice or currently under development lost all their antiviral activity against Omicron. The situation also became increasingly complex as several subvariants (BA.1, BA.2, etc.) emerged over the following weeks. So the scientists tried to determine whether the monoclonal antibodies were effective against the new strains that were co-circulating. In a study published in March 2022, they provided a number of answers. The casirivimab/imdevimab combination, which until then had offered a similar level of protection against severe forms of COVID-19 as vaccination, was no longer active against Omicron. The neutralizing activity of another antibody cocktail (tixagevimab/cilgavimab) was significantly reduced against BA.1 compared with the Delta variant, but its neutralizing activity was not reduced nearly as much against BA.2. This result, which showed that a monoclonal antibody could lose its neutralizing capability for a given variant before recovering it for a subsequent variant, emphasizes the importance of continuing with efficacy testing on all available antibodies (Figure 1).

The protective efficacy of monoclonal antibodies depends primarily, but not solely, on their neutralizing capability. Some monoclonal antibodies can also induce a key immune defense mechanism known as antibody-dependent cellular cytotoxicity (ADCC), which kills cells infected with SARS-CoV-2. In the case of COVID-19, natural killer (NK) cells in the immune system recognize the antibodies that bind to antigens on the surface of infected cells and then specifically lyse those cells. These antibodies are described as "polyfunctional." This characteristic is all the more interesting since the monoclonal antibodies with the highest neutralizing capability are not those that induce ADCC the most, as shown in a study published in December 2022. In short, to obtain protective antibody cocktails, it may be a good idea to include polyfunctional antibodies which combine a high neutralizing capability with a strong ability to induce immune defense mechanisms [12].

Lastest Monoclonal Antibodies for Pre-Exposure Prophylaxis in Patients for the most vulnerable persons at June 2024

Pemivibart (Pemgarda): The FDA issued an emergency use authorization (EUA) for pemivibart (Pemgarda) opens in a new tab or window as COVID-19 pre-exposure prophylaxis in immunocompromised individuals who are unlikely to mount a sufficient immune response following vaccination. A long-acting monoclonal antibody, pemivibart is specifically authorized for people ages 12 years and older (and weighing 40 kg or more) with moderate-to-severe immune compromise either because of a medical condition or due to immunosuppressant medications. Pemivibart is given as a single intravenous infusion and is not for use as post-exposure prophylaxis or in people currently infected with SARS-CoV-2.

The EUA was based on immunobridging data involving other human monoclonal antibodies against SARS-CoV-2 demonstrating that pemivibart may be effective for COVID prevention. Serum neutralizing antibody titers of Pemgarda were consistent with the titer levels associated with efficacy in prior clinical trials of adintrevimab and certain other monoclonal antibody products.

According to the EUA, individuals who would qualify for the antibody include those undergoing active treatment for cancer (including those receiving CAR T-cell therapy or stem cell transplant); patients with hematologic malignancies associated with poor responses to COVID vaccines regardless of their treatment status; solid-organ transplant recipients; those with moderate-or-severe primary immunodeficiency; people with advanced or untreated HIV; and those on high-dose corticosteroids, B-cell depleting agents, and other immunosuppressants.

No long-acting monoclonal antibody has been available for preventing COVID infection in individuals with moderate-to-severe immune compromise since the agency pulled the EUA opens in a new tab or window for tixagevimab-cilgavimab (Evusheld) in January 2023 -- the move followed data showing the combination was unlikely to be sufficiently active against circulating SARS-CoV-2 variants. At the time, the CDC recommended opens in a new tab or window that immunocompromised individuals receive the latest COVID booster (if they had not already), wear a well-fitting high-quality mask in public, maintain distance in crowded areas, and improve indoor ventilation.

Pemivibart is administered at a dose of 4,500 mg over a 60-minute infusion, with repeat dosing every 3 months recommended if ongoing protection is needed. FDA cautioned that anaphylaxis occurred in 0.6% of clinical trial participants who received pemivibart. Therefore, patients should be monitored for 2 hours after the infusion is finished, and pemivibart should be administered in settings where health providers have immediate access to medications to reverse severe allergic reactions and can alert EMS if necessary. Other potential side effects noted in the labelingopens in a new tab or window include infusion-related reactions, fatigue, nausea, and headache [13].

SUPERNOVA Phase III trial of sipavibart long-acting antibody met primary endpoints in preventing COVID-19 in immunocompromised patient population

SUPERNOVA is a large Phase III global trial providing the only efficacy data in immunocompromised patients, demonstrating the potential benefit of a COVID-19 antibody against recent SARS-CoV-2 variants. Immunocompromised patients include those with blood cancer, organ transplant recipients, patients with end-stage renal disease requiring dialysis, patients receiving B-cell depleting therapy within the past year, and those taking immunosuppressive medications. Despite accounting for approximately 4% of the population, immunocompromised patients make up about 25% of COVID-19 hospitalisations, ICU admissions, and deaths, even after multiple doses of COVID-19 vaccines [14].

The trial met both dual primary endpoints; the first one being the relative risk reduction of symptomatic COVID-19 caused by any SARS-CoV-2 variant and the second being the relative risk reduction of infections caused by SARS-CoV-2 variants not containing the F456L mutation. SUPERNOVA demonstrated the potential benefit of sipavibart in an evolving variant landscape in which COVID-19 cases captured over the course of the trial were caused by several different SARS-CoV-2 variants.

Primary efficacy endpoints. The first evaluated the efficacy of sipavibart against any confirmed SARS-CoV-2 positive symptomatic illness occurring post dose prior to day 181 caused by any variant (i.e., all cases regardless of if the variant has the F456L mutation or not, which sipavibart is not expected to neutralise). The second dual primary efficacy analysis was conducted using only the confirmed COVID-19 cases in the trial where the variant causing the COVID-19 cases did not have the F456L mutation, referred to as a “matched” variant analysis.

Participants were individuals 12 years of age and over who would benefit from prevention with the investigational LAAB, defined as having increased risk for inadequate response to active immunisation (predicted poor responders to vaccines or intolerant of vaccine). Participants at the time of screening had a negative point-of-care SARS-CoV-2 serology test. Participants will be followed for 15 months, with SARS-CoV-2 neutralising antibodies assessed at one, three and six months.

All participants in the trial had an immunocompromising condition and/or were on immunosuppressive treatments, which put them at risk to mount an inadequate immune response to vaccination and at high risk of developing severe COVID-19. This included patients with hematologic malignancies, solid organ transplant recipients, hematopoietic stem cell transplants, end stage kidney disease/dialysis and being within one year of receipt of B cell depleting therapy, among others. Across the treatment groups, demographic and baseline characteristics were generally well balanced.

Positive high-level results from the SUPERNOVA Phase III COVID-19 pre-exposure prophylaxis (prevention) trial showed AstraZeneca’s sipavibart (formerly AZD3152), an investigational long-acting antibody (LAAB), demonstrated a statistically significant reduction in the incidence of symptomatic COVID 19 compared to control (tixagevimab/cilgavimab or placebo) in an immunocompromised patient population.

COVID-19 still represents a significant and disproportionate risk for immunocompromised patients, with infection often leading to serious and protracted illness.

Infection-fighting antibodies directly to patients who often don’t respond adequately to vaccines, the data support that sipavibart has the potential to provide much-needed protection against COVID-19 in this highly vulnerable population [15,16].

Immunocompromised patients currently have limited or no options for COVID-19 protection and continue to face a significant burden of disease, despite often being fully vaccinated. Sipavibart has the potential to prevent COVID-19 in the immunocompromised and we will now work with regulatory authorities globally to bring sipavibart to these vulnerable patients.

Sipavibart was well tolerated in the trial and preliminary analyses show adverse events were balanced between the control and sipavibart arms. The data will be presented at a forthcoming medical meeting. AstraZeneca is in dialogue with regulatory authorities on potential authorisation or approval pathways [17-19].

Sipavibart

Sipavibart (formerly AZD3152) is an investigational long-acting monoclonal antibody (LAAB) against COVID-19. Sipavibart was designed to provide broad and potent coverage across Omicron and ancestral viral variants by neutralising spike protein interaction with the host receptor ACE2.

Sipavibart was derived from B-cells donated by convalescent patients after SARS-CoV-2 infection. Sipavibart has been engineered using the same antibody scaffold as Evusheld and was optimized with the same half-life extension and reduced Fc effector function and complement C1q binding platform. The reduced Fc effector function aims to minimise the risk of antibody-dependent enhancement of disease - a phenomenon in which virus-specific antibodies promote, rather than inhibit, infection and/or disease. Sipavibart was licensed by AstraZeneca in May 2022 from RQ Biotechnology [20].

Patients Immunocompromysed and Sars-Cov-2

Individuals with immunosuppressed conditions, including people with primary immunodeficiencies and secondary immunodeficiencies, consisting of people with solid-organ trans-plants, metastatic cancers, hematologic malignancies, advanced or untreated HIV (human immunodeficiency virus) infection, those receiving cancer chemotherapy, and patients with autoimmune diseases receiving immunosuppressive biologics and medications.

Patients in this heterogenous group had a higher risk of COVID-19-related hospitalization, severe COVID-19, or death and tend to have higher risk for opportunistic infections.  In addition, prolonged SARS-CoV-2 infection and persistent viral replication in ICP notonly cause long duration of symptoms but also risk of emergence of antiviral-resistant or vaccine-escaped variants, prolonging the pandemic.

Although COVID-19 treatment guidelines had been proposed to manage patients with different severities and clinical cohorts, a consensus on COVID-19 patients in ICP was lacking and the information was limited.

Immunocompromised individuals are at higher risk of severe COVID-19 outcomes.

Vaccination remains a critical preventive mea-sure for this vulnerable population. Although some may have a reduced response to vaccines, receiving the recommended doses can still provide some level of protection and potentially mitigate severe disease.

Given the potential for diminished vaccine response, immunocompromised individuals should be considered for booster doses based on local guidelines and emerging data.   In cases of exposure to COVID-19, immunocompromised individuals may require specific isolation or quarantine measures, depending on their risk profile. The management of COVID-19 in ICP necessitates an individualized approach. Clinicians must carefully consider the patient's specific immunocompromised condition, medical history, and risk factors to determine the most appropriate treatment plan. In severe COVID-19 cases among ICP, antiviral therapies such as remdesivir may be considered.

The decision to use antiviral drugs should be based on clinical judgment and consultation with specialists from multidisciplinary areas.

In severe COVID-19 cases with significant inflammatory responses, corticoste-roids like dexamethasone may be used under close medical supervision. The use of CCP therapy in specific severe cases of COVID-19 among immunocompromised patients may be considered on a case-by-case basis early in the pandemic.

Therapeutic recommendations for antiviral or immunomodulator therapy of adults with varying severities of COVID-19 are summarized in (Table 1).

Effectiveness of Monoclonal Antibody-based therapy Against Covid Variants (June 2024)

Monoclonal antibodies against the SARS-CoV-2 S protein act through mechanisms related to their structure. First, the antigen-binding fragments (Fab) prevent the virus from binding to the ACE2 receptors, and second, the Fc fragment can activate the complement system and bind to the Immunoglobulin Fc receptors (FcRs) on cytotoxic cells that can eliminate virus-infected cells through Ab-dependent cell-mediated cytotoxicity (ADCC). Unfortunately, some mAbs can bind to macrophage FcRs and induce a hyperinflammatory response resulting from Ab-dependent enhancement (ADE) of cytokine production.

The SARS-CoV-2 RBD has become the main target of mAbs because of its crucial role in virus entry into host cells (Table 2). Analysis of the structural relationship between RBD and anti-RBD NAbs has led to the classification of these antibodies according to structural features and mechanism of action. Class 1 NAbs, e.g., CT-P59 (regdanvimab), target the receptor binding motif (RBM). They recognize the RBD in the up conformation, thus blocking the interaction with the ACE2 receptor. Class 2 NAbs, e.g. LY-CoV1404 (bebtelovimab), target the ACE2 binding site of the RBD in both up and down conformations. Class 3 antibodies, e.g., S309 (sotrovimab), target the conserved core domain of the RBD without altering interactions with the ACE2 receptor. Class 4 antibodies, e.g., S2X259, target epitopes in both the RBM and the core domain of the RBD. Unfortunately, frequent mutations in the RBD have modified the epitopes recognized by mAbs, resulting in the emergence of viral variants resistant to mAbs. To address this issue, researchers are exploring other SRS-CoV-2 regions as potential targets for therapeutic mAbs.

Monoclonal Antibodies Aprovation as Prophylaxis-Therapy in the Ederly and Immonocompromised Sars-Cov-2 population at June 2024

Currently, most mAbs are ineffective at providing an immune response to Omicron strains post BA.2. Recently, the US Food and Drug Administration and provinces in Canada have found tixagevimab plus cilgavimab ineffective against Omicron variants [21].

Similar decisions in the US have been made previously for bamlanivimab monotherapy, which was revoked in April 2021 because of low efficacy against newer COVID-19 variants [22].

 In the context of increasing prevalence of resistant SARS-CoV-2 subvariants, the decision to administer tixagevimab plus cilgavimab, or any other mAbs to a given patient should be based on regional prevalence of resistant variants, individual patient risks, available resources, and logistics. Further, patients who receive mAbs as a prophylactic for COVID-19 should continue taking precautions, including proper hand hygiene, physical distancing, and mask wearing to avoid exposure (Table 3).

Although mAbs demonstrated effectiveness, concerns have been raised regarding the potential for creating spike protein resistance-associated viral mutations, particularly in immunocompromised patients.

A study conducted from January to February 2022 investigated whether resistance-associated mutations developed after treatment with sotrovimab in high-risk patients. Out of the high-risk patients, specimens were collected at three time points from 14 of the 18 patients (78%). Genomic analysis revealed that all 18 (100%) patients were infected with the Omicron variant; 17 with BA.1 (94%) and one with BA.2 (6%). Ten patients (56%) developed receptor-binding domain mutations at spike position E340 or P337 within 3-31 days after treatment. The researchers identified six mutations in the spike protein S: E340K/A/V/D/G/Q and three in S: P337L/R/S. Mutations increased over time, exceeding 50% between days 5 and 28. Patients with mutations had significantly delayed time to viral clearance (mean, 32 [SD, 8.1] days vs 19.6 [SD, 11.1] days for those without mutations; HR, 0.11 [95% CI, 0.02-0.60]). No S: E340 or S: P337 mutations were found in the Omicron variant from sequences in the general population. The four patients with the sotrovimab resistance-associated S: E340K mutation were immunocompromised [23].

Evidence of how Fc-dependent antibody functions may impact infection consequences within immunocompromised populations is still limited, requiring a more robust framework for evaluation.

Sotrovimab is one of the few mAbs that demonstrated retained favorable clinical outcomes against the Omicron variant and as such it is crucial to understand Fc-mediated effects in order to evaluate and improve application of antibody therapy.

The Omicron variant presents a heightened risk to patients that are immunocompromised due to their inability to mount a sufficient antibody response, even when they are vaccinated and/or have previous COVID-19 infections. This reality places immunocompromised patients at risk of death and hospitalization due to increased likelihood of high viral load and their difficulty in eliminating the virus. There is a continued need for research supporting multiple COVID-19 prophylaxis. The medical and scientific community can best serve their immunocompromised patients by updating their understanding of COVID-19 prophylaxis and its utility in supporting immunocompromised patients. Moreover, there is an urgent need for new randomized controlled trials in vaccinated, immunocompromised subjects, during current strains of COVID-19 to support the development of more effective mAbs (Table 4).

SARS-CoV-2 variants of concern as of 26 June 2024

New evidence is regularly assessed on variants detected through epidemic intelligence, rules-based genomic variant screening, or other scientific sources. If a decision is made to add, remove, or change the category for any variant, the tables are updated to reflect this change. The tables are regularly sent for consultation to ECDC and WHO Regional Office for Europe’s joint virus characterisation working group.

Variant classification serves as an important communication tool for alerting EU/EEA countries about the emergence of SARS-CoV-2 variants with concerning properties likely to impact the epidemiological situation in the EU/EEA.

ECDC utilises three categories of variant classification to communicate increasing levels of concern about a new or emerging SARS-CoV-2 variant: variant under monitoring (VUM), variant of interest (VOI) and variant of concern (VOC). Classification criteria and recommended Member state actions are available here: ECDC variant classification criteria and recommended Member State actions.

New evidence is regularly assessed on variants detected through epidemic intelligence, genomic horizon scanning, or other scientific sources. If a decision is made to add, remove, or change the category for any variant, the tables are updated to reflect this change. The tables are regularly sent for consultation to ECDC and WHO Regional Office for Europe’s joint virus characterisation working group.

Variant surveillance data, including the distribution of VOC and VOI variant proportions in the EU/EEA, is presented as part of the European Respiratory Virus Surveillance Summary (ERVISS).

Description of the tables

Category: variant of concern (VOC), variant of interest (VOI), or variant under monitoring (VUM).

WHO label: As of 31st May 2021, WHO proposed labels for global SARS-CoV-2 variants of concern and variants of interest to be used alongside the scientific nomenclature in communications about variants to the public. This list includes variants on whose global list of VOC and VOI, and is updated as WHO’s list changes.

Lineage and additional mutations: the variant designation specified by one or more Pango lineages and any additional characteristic spike protein changes. An alternate description may be used if the variant is not easy to describe using this nomenclature. For updated information on Pango lineages and definition of lineages and for instructions on how to suggest new lineages, visit the Pango lineages website. Each lineage in then table is linked to the respective lineage page on the Pango lineages website.

Country first detected: only present if there is moderate confidence in the evidence relating to the first country of detection.

Spike mutations of interest: not all spike protein amino acid changes are included – this is not a full reference for assignment of the variants. It includes changes to spike protein residues 319-541 (receptor binding domain) and 613-705 (the S1 part of the S1/S2 junction and a small stretch on the S2 side), and any additional unusual changes specific to the variant.

Year and month first detected: as reported in the GISAID EpiCoV database. This can be adjusted backwards in time if new retrospective detections are made.

Evidence concerning properties in three different categories:

·       Transmissibility

·       Immunity

·       Infection severity

Each category is annotated as increased, reduced, similar, unclear, or no evidence depending on the currently available evidence. Increased or reduced means that there is evidence demonstrating that the property is different enough for the variant compared to previously circulating variants that it is likely to have an impact on the epidemiological situation in the EU/EEA. Similar means that there is evidence that demonstrates that the property is not different enough for this variant compared to previously circulating variants that it is unlikely to have an impact. Unclear means that the current evidence is preliminary or contradictory enough to make the assessment uncertain. No evidence means that no evidence has yet been evaluated for this category. The evidence is further annotated with v or m to indicate whether the evidence is available for the variant itself (v) or for mutations associated with the variant (m).

Transmission in the EU/EEA: categorised as dominant, community, outbreak(s), and sporadic/travel. The categories are qualitative, and the assessment is based on surveillance data collected in TESSy, GISAID EpiCoV data, epidemic intelligence data, and direct communications with the affected countries.

 Variants of Concern (VOC)

As of 3 March 2023, ECDC has de-escalated BA.2, BA.4 and BA.5 from its list of SARS-CoV-2 variants of concern (VOC), as these parental lineages are no longer circulating. ECDC will continue to categorise and report on specific SARS-CoV-2 sub-lineages in circulation that are relevant to the epidemiological situation. There are currently no SARS-CoV-2 variants meeting the VOC criteria.

Reported protection and antibody concentration from RCTs of monoclonal antibodies in preventing COVID-19. Searched MEDLINE, PubMed, Embase, and the Cochrane COVID-19 Study Register for randomized placebo-controlled trials of SARS-CoV-2-specific monoclonal antibodies (mAbs) used as pre-exposure and peri-exposure prophylaxis for COVID-19. They was included only studies where both protection from symptomatic infection and pharmacokinetic information of the monoclonal antibody were provided within the same study. They was identified six eligible studies assessing monoclonal antibodies as pre-exposure and peri-exposure prophylaxis for COVID-. The antibodies used in these studies were casirivimab/imdevimab (three studies), bamlanivimab, cilgavimab/tixagevimab, and adintrevimab. Omicron variants were the dominant circulating variants. One study assessed protection in two time periods; firstly in a pre-Omicron period when the Delta variant was the dominant circulating variant, and separately later when Omicron variants BA.1 and BA.1.1 were the dominant variants13. The overall efficacies against pre-Omicron variants in the included studies ranged from 68.6% to 92.4%. Stadler et al. was identified a trend for lower efficacies with increasing time since administration and against the escaped variant, the latter being reported previously by Schmidt et al. [24-27] (Figure 2).

The efficacy at each time interval is shown in blue (points indicate observed efficacy, horizontal error bars indicate time interval and vertical error bars represent 95% CIs of efficacy). The antibody concentration is shown in black. a Antibody concentration (n?=?1776 individuals) and efficacy data (n?=?5172 individuals) for cilgavimab/tixagevimab was extracted from Levin et al.13 b Single administration of casirivimab/imdevimab data are a combination of data from O’Brien et al.14 and Herman et al.15 who report on the same clinical trial over different follow-up intervals. Efficacy data were reported weekly over the first four weeks in O’Brien et al. (diamonds) (n?=?1505), and monthly for eight months in Herman et al. (circles) (n?=?1683). Antibody concentration data was reported up to day 168 in O’Brien et al. (solid line, b n?=?12), and modeling of the pharmacokinetic profile of the antibody concentration, reported in Herman et al., was used to inform the antibody concentration between 168 and 240 days (dashed line, b). c Isa et al.16 reported efficacy (n?=?969) and in vivo concentration after repeated administration of 1.2?g of casirivimab/imdevimab every 4 weeks (n?=?723). Hence, the antibody concentration did not decline as in the other studies. d The modeled concentration of adintrevimab after a single administration was extracted from the study by Schmidt et al.12. The efficacy of adintrevimab was reported both when the delta variant was dominant (circles) (n?=?1267) and when Omicron variants BA.1 and BA.1.1 were dominant (triangles) (n?=?378).

Conclusion

COVID-19 still represents a significant and disproportionate risk for immunocompromised patients, with infection often leading to serious and protracted illness. Infection-fighting antibodies directly to patients who often don’t respond adequately to vaccines, the data support that sipavibart has the potential to provide much-needed protection against COVID-19 in this highly vulnerable population. Immunocompromised patients currently have limited or no options for COVID-19 protection and continue to face a significant burden of disease, despite often being fully vaccinated. Sipavibart has the potential to prevent COVID-19 in the immunocompromised and we will now work with regulatory authorities globally to bring sipavibart to these vulnerable patients.

Studying the adaptation trajectory of SARS-CoV-2, it is crucial to anticipate possible future events rooted in the molecular mechanisms that underpin the evolutionary success of SARS-CoV-2 is essential. The potential role of advanced Treatment as Pre-Exposure Prophylaxis against SARS-CoV-2 has introduced it as a new platform to encourage the adaptation of emerging medical technologies for infectious diseases.

The use of monoclonal antibodies for Pre-Exposure in these cohorts has the potential to provide long-term protection from both symptomatic and severe COVID-19 for these vulnerable groups. However, the frequent observation of novel SARS-CoV-2 variants that escape antibody recognition has raised significant challenges in predicting monoclonal antibody protection against new variants. Several studies have investigated the efficacy of monoclonal antibodies as pre- and post-prophylaxis for COVID-19. Historical evidence is promising; however, new variants of concern are proving challenging for currently available regimens.


References

1.      Vardavas CI, Mathioudakis AG, Nikitara K, Stamatelopoulos K, Georgiopoulos G, Phalkey R, et al. Prognostic factors for mortality, intensive care unit and hospital admission due to SARS-CoV-2: a systematic review and meta-analysis of cohort studies in Europe. Euro Respiratory Rev. 2022; 31.

2.      Ryan L. Vaccinated but Not Protected - Living Immunocompromised During the Pandemic. JAMA. 2021; 325: 2443-2444.

3.      Herman GA, O'Brien MP, Forleo-Neto E, Sarkar N, Isa F, Hou P, et al. Efficacy and safety of a single dose of casirivimab and imdevimab for the prevention of COVID-19 over an 8-month period: a randomised, double-blind, placebo-controlled trial. The Lancet Infectious Diseases. 2022; 22: 1444-1454.

4.      Levin MJ, Ustianowski A, De Wit S, Launay O, Avila M, Templeton A, et al. Intramuscular AZD7442 (tixagevimab–cilgavimab) for prevention of Covid-19. New England J Medicine. 2022; 386: 2188-200.

5.      Cao Y, Yisimayi A, Jian F, Song W, Xiao T, Wang L, et al. BA.2.12.1, BA.4 and BA.5 escape antibodies elicited by Omicron infection. Nature. 2022; 608: 593-602.

6.      Yamasoba D, Kosugi Y, Kimura I, Fujita S, Uriu K, Ito J, Sato K. Neutralisation sensitivity of SARS-CoV-2 omicron subvariants to therapeutic monoclonal antibodies. The Lancet Infectious Diseases. 2022; 22: 942-943.

7.      United States Food Drug Administration. 2022.

8.      Abdelnabi R, Foo CS, Kaptein SJ, Zhang X, Do TN, Langendries L, et al. The combined treatment of Molnupiravir and Favipiravir results in a potentiation of antiviral efficacy in a SARS-CoV-2 hamster infection model. eBioMedicine. 2021; 72: 103595.

9.      Weinreich DM, Sivapalasingam S, Norton T, Ali S, Gao H, Bhore R, et al. REGN-COV2, a neutralizing antibody cocktail, in outpatients with Covid-19. New England J Medicine. 2021; 384: 238-251.

10.    World Health Organization (2022). Therapeutics and COVID-19: living guideline. 2022.

11.    Van Erp EA, Luytjes W, Ferwerda G, Van Kasteren PB. Fc-mediated antibody effector functions during respiratory syncytial virus infection and disease. Frontiers in immunology. 2019; 10: 548.

12.    Evans RA, Dube S, Lu Y, Yates M, Arnetorp S, Barnes E, et al. Impact of COVID-19 on immunocompromised populations during the Omicron era: insights from the observational population-based INFORM study. The Lancet Regional Health–Europe. 2023; 35.

13.    Dube S. Continued Increased Risk of COVID-19 Hospitalisation and Death in Immunocompromised Individuals despite Receipt of ?4 Vaccine Doses: Updated 2023 Results from INFORM, a Retrospective Health Database Study in England.

14.    Turtle L. Individuals with Multiple Sclerosis Are at High Risk for COVID-19 Hospitalisation and Death Despite High Rates of Vaccination: Results from the England INFORM Study. Oral Presentation at ECCMID 2024.

15.    Meeraus W. High Prevalence of Immunocompromising Conditions among Patients with Severe Acute Respiratory Infection, Including SARS-CoV-2: Results from a Multicentre, Test-Negative Case Control Study. Abstract #01796 at ECCMID 2024.

16.    Meeraus W. Immunocompromise, Cancer and Other Comorbidities in Patients with Severe Acute Respiratory Infection Testing Positive versus Negative for SARS-CoV-2: A Post Hoc Analysis of COVIDRIVE Data from May 2021 to May 2023. Abstract #01800 at ECCMID 2024.

17.    Ketkar A, Willey V, Pollack M, Glasser L, Dobie C, Wenziger C, et al. Assessing the risk and costs of COVID-19 in immunocompromised populations in a large United States commercial insurance health plan: the EPOCH-US Study. Current medical research and opinion. 2023; 39: 1103-1118.

18.    Miljanovic D, Cirkovic A, Lazarevic I, Knezevic A, Cupic M, Banko A. Clinical efficacy of anti-SARS-CoV-2 monoclonal antibodies in preventing hospitalisation and mortality among patients infected with Omicron variants: a systematic review and meta-analysis. Rev Med Virol. 2023; 2439

19.    https://www.cbc.ca/news/canada/calgary/alberta-covid-therapy-evusheld-1.6731381

20.    Francica JR, Cai Y, Diallo S, Rosenthal K, Ren K, Flores DJ, Dippel A, et al. The SARS-CoV-2 Monoclonal Antibody AZD3152 Potently Neutralizes Historical and Emerging Variants and is Being Developed for the Prevention and Treatment of COVID-19 in High-risk Individuals. InOpen Forum Infectious Diseases. 2023; 10.

21.    Cowan J, Amson A, Christofides A, Chagla Z. Monoclonal antibodies as COVID-19 prophylaxis therapy in immunocompromised patient populations. Int J Infectious Diseases. 2023; 134: 228-238.

22.    Stadler E, Burgess MT, Schlub TE, Khan SR, Chai KL, McQuilten ZK, et al. Monoclonal antibody levels and protection from COVID-19. Nature Communications. 2023; 14: 4545.

23.    Schmidt P, Narayan K, Li Y, Kaku CI, Brown ME, Champney E, et al. Antibody-mediated protection against symptomatic COVID-19 can be achieved at low serum neutralizing titers. Science Translational Medicine. 2023; 15: 2783.

24.    Levin MJ, Ustianowski A, De Wit S, Launay O, Avila M, Templeton A, et al. Intramuscular AZD7442 (tixagevimab–cilgavimab) for prevention of Covid-19. New England J Med. 2022; 386: 2188-200.

25.    O’Brien MP, Forleo-Neto E, Musser BJ, Isa F, Chan KC, Sarkar N, et al. Subcutaneous REGEN-COV antibody combination to prevent Covid-19. New England J Medicine. 2021; 385: 1184-1195.

26.    Herman GA, O'Brien MP, Forleo-Neto E, Sarkar N, Isa F, Hou P, et al. Efficacy and safety of a single dose of casirivimab and imdevimab for the prevention of COVID-19 over an 8-month period: a randomised, double-blind, placebo-controlled trial. The Lancet Infectious Diseases. 2022; 22: 1444-1254.

27.    COVID-19: which monoclonal antibodies should be used for vulnerable individuals?. Institut Pasteur/Olivier Schwartz & Félix Rey. 2024.