910
Views
0
CrossRef citations to date
0
Altmetric
Special Report

Monoclonal antibodies for the treatment of COVID-19 infection in children

&
Pages 1529-1535 | Received 24 Aug 2022, Accepted 06 Oct 2022, Published online: 19 Oct 2022

ABSTRACT

Introduction

Monoclonal antibodies (mAbs) have been authorized for the treatment of COVID-19 in pediatric populations, however, there is a lack of evidence for their use in these populations.

Areas covered

We outline the evidence of mAbs for COVID-19, discuss their use in the treatment of COVID-19 infection for pediatric patients, and consider alternative treatment options and challenges to COVID-19 drug approvals.

Expert opinion

Limited evidence exists for the safety and efficacy of mAbs to treat COVID-19 in children as new variants emerge. In rare pediatric outpatient settings, such as profound immunodeficiency or severe pulmonary disease, the benefits of antiviral treatment for COVID-19 likely outweigh the relatively small risks. However, for the great majority of pediatric patients, mAb treatment is likely not indicated. Small molecule antiviral therapies are another potential treatment for COVID-19 in children in an outpatient setting, though neither mAb nor small molecule antiviral treatments have significant supporting evidence in children and developing a strong evidence base for these decisions will be challenging if not impractical. Ultimately, these decisions are likely to be made at the level of individual cases using expert opinion as the primary guiding principle.

1. Introduction

Coronavirus disease (COVID-19) caused by the SARS-CoV-2 virus remains a threat to human health worldwide, with multiple variants evolving over recent years, most recently including the highly infectious Omicron variant and its sub-variants.

Vaccination is the most effective strategy to prevent infection and to slow transmission of COVID-19 [Citation1–3]. In the United States, four COVID-19 vaccines have been approved or authorized by the Food and Drug Administration (FDA) for emergency use as of August 2022: Ad26.COV2, NVX-CoV2373, BNT162b2, and mRNA-1273 [Citation4–10] . However, the efficacy of currently approved or authorized vaccines has waned since Omicron has emerged as the dominant variant, necessitating the development of therapies to treat infection [Citation11,Citation12].

Passive humoral immunotherapy with neutralizing monoclonal antibodies (mAbs) has emerged as a potential treatment option to prevent hospitalization and death related to COVID-19 [Citation13,Citation14]. In recognition of this, the mAb, bebtelovimab, remains authorized by the FDA for emergency use in children and adults to prevent severe disease [Citation15,Citation16]. Several additional mAbs that were authorized earlier in the pandemic have lost authorization due to the inability to bind the omicron variant [Citation17–20]. For each of these antibodies, a paucity of evidence exists for use in pediatric populations.

In this review, we will outline the evidence of mAbs for COVID-19 treatment, examine their use in treating pediatric patients with COVID-19 infection, and discuss alternative treatment options and challenges to COVID-19 drug approvals.

2. Anti-Sars-CoV-2 monoclonal antibodies

2.1. Bamlanivimab-etesevimab

The FDA granted emergency use authorization (EUA) for bamlanivimab-etesevimab, administered together, in February 2021. The authorization was based on analysis of 1035 unvaccinated ambulatory patients with mild-to-moderate COVID-19 at risk for progression to severe disease enrolled from September – December 2020, when Alpha was the predominant variant of concern (VOC) [Citation18]. The study includes 11 adolescents age 12–17 years. At day 29, 11/518 (2.1%) patients in the treatment arm vs 36/517 (7%) in the placebo arm had a COVID-19-related hospitalization or death, corresponding to a 70% relative risk reduction [Citation18]. No deaths occurred in the treatment group, while 10 deaths occurred in the placebo group. All deaths occurred in adults. Serious adverse events (SAEs) were rare [Citation18].

Bamlanivimab-etesevimab effectively neutralizes the Wuhan strain of SARS-CoV-2 and the alpha and delta variants but has reduced activity against gamma and lost activity against the Omicron variants [Citation21,Citation22]. Because the Omicron BA.5 subvariant is now the dominant circulating subvariant in the United States as of August 2022, the FDA has limited use of this combination.

2.2. Sotrovimab

In May 2021, the FDA granted emergency use authorization (EUA) for sotrovimab based on an interim analysis of 583 unvaccinated, non-hospitalized adults with mild-to-moderate COVID-19 enrolled between August 2020 – March 2021, when Alpha was the predominant VOC [Citation20]. Hospitalization or death occurred in 21/292 (7%) patients who received placebo versus 3/291 (1%) patients treated with sotrovimab, an 85% relative risk reduction [Citation20]. Adverse events (AEs) attributable to sotrovimab were rare [Citation20,Citation23].

Sotrovimab retains in-vitro activity against the Omicron BA.1 and BA.1.1 subvariants, but it has substantially decreased in-vitro activity against Omicron BA.2, BA.4 and BA.5 and is not expected to provide clinical benefit for patients infected with these variants. Because of this, the FDA has discontinued the authorization for sotrovimab.

2.3. Casirivimab-imdevimab (REGEN-COV)

The efficacy of casirivimab-imdevimab for COVID-19 treatment was evaluated in a phase 3 placebo-controlled study of 4057 unvaccinated participants [Citation24]. The efficacy analysis included SARS-CoV-2 positive participants aged ≥18 years who had ≥1 risk factor for progression to severe COVID-19, enrolled between September 2020 – January 2021 when Alpha predominated [Citation19]. For the participants who received 1200 mg, COVID-19 related hospitalization or death occurred in 7/736 (1%), compared to 24/748 (3.2%) of those who received placebo- a 70.4% relative risk reduction. For patients who received 2400 mg of casirivimab-imdevimab, hospitalization or death occurred in 18/1355 (1.3%) versus 62/1341 (4.6%) of patients in the placebo group [Citation24].

Casirivimab-imdevimab also lost neutralizing activity against the omicron variant, and the FDA limited its use only to regions where there is likely a susceptible variant [Citation21,Citation25].

2.4. Bebtelovimab

As of August 2022, bebtelovimab is the only FDA-authorized mAb currently used in the United States for COVID-19 treatment as it is the only therapeutic antibody that retains full activity against the omicron variants in neutralization assays [Citation15,Citation16,Citation26]. The data supporting the EUA are based on analysis of a portion of a phase 2 trial that enrolled 714 patients with mild-to-moderate COVID-19 between May and July 2021 when Alpha and Delta variants were predominant [Citation15]. Those at low-risk for severe COVID-19 were unvaccinated and randomized 1:1:1 to placebo, bebtelovimab 175 mg, or bebtelovimab 175 mg + bamlanivimab 700 mg + etesevimab 1400 mg. High-risk adults and pediatric subjects (≥12 years weighing ≥40 kg), 31% of whom were vaccinated at baseline, received open-label active treatments of bebtelovimab or bebtelovimab + bamlanivimab + etesevimab. The primary endpoint was the proportion of subjects with persistently high viral load (PHVL) by Day 7. Of the low-risk patients, PHVL occurred in 25/126 subjects treated with placebo (19.8%), compared to 15/125 (12%) subjects treated with bebtelovimab alone, and 16/126 (12.7%) subjects treated with bebtelovimab + bamlanivimab + etesevimab. Although the rate of PHVL decreased, this finding did not reach a level of statistical significance (p = 0.097 BEB vs placebo; p = 0.132 bebtelovimab + bamlanivimab + etesevimab vs placebo) [Citation15]. The key secondary endpoint of viral load area under the curve was reduced in low-risk patients treated with bebtelovimab compared to placebo (p = 0.006).

In the high-risk arm, the primary endpoint was safety. AEs occurred in 37/380 (9.7%) of low-risk and 48/326 (14.7%) of high-risk patients treated with bebtelovimab or bebtelovimab + bamlanivimab + etesevimab; the majority of AEs were considered mild or moderate in severity. SAEs were reported in 7/326 (2.1%) of high-risk patients, including one death (a cerebrovascular accident); 1 SAE was reported among low-risk patients [Citation15].

2.5. Tixagevimab-cilgavimab (EVUSHELD)

In February 2022, The FDA granted EUA for Tixagevimab-cilgavimab for individuals ≥12 years weighing ≥40 kg who may not have an adequate response to COVID-19 vaccination or who cannot receive a full series of a COVID-19 vaccine [Citation27]. Tixagevimab-cilgavimab has retained some activity against omicron subvariants and is authorized for the pre-exposure prophylaxis of COVID-19, but is not approved for COVID-19 treatment [Citation25].

3. Small molecule antiviral therapies for SARS-CoV-2

Two of the three small molecule antiviral therapies that have been authorized or approved for use in treating COVID-19 are available to pediatric patients as of August 2022. Remdesivir, an inhibitor of viral RNA-dependent RNA polymerase with the ability to inhibit SARS-CoV-2 in vitro, is approved to treat pediatric patients ≥28 days of age weighing ≥3 kg [Citation28–30]. Nirmatrelvir with Ritonavir (Paxlovid) is an oral protease inhibitor that has received EUA for adults and children age ≥12 years of age weighing ≥88 lbs [Citation31]. Molnupiravir, a nucleoside analogue that inhibits viral replication, is only approved in adults [Citation32].

4. Choice of antiviral therapies

Small molecule antivirals and anti-SARS-CoV-2 mAbs both show strong efficacy against COVID-19, but with different benefits and limitations. The small molecule antivirals described above target the active sites of highly conserved viral proteins that have proved resistant to mutation, allowing for use across multiple viral variants [Citation33]. In vitro analysis shows that these antivirals have retained their activity against all variants of concern including Omicron [Citation33]. The mAbs target the highly variable spike protein, recognizing a limited peptide sequence on its surface, and have lost efficacy with the emergence of new variants [Citation21,Citation25]. This is not surprising as the virus is under constant selective pressure to evade the human immune response in each productive infection, the same immune response that was leveraged to identify therapeutic antibodies. In all likelihood, future variants of concern will continue to evade mAb therapeutics while displaying relative sensitivity to the small molecule antivirals.

Although SARS-CoV-2 will likely continue to mutate to evade mAbs [Citation21,Citation25], generating antigen-specific mAbs for commercial use can be done rapidly [Citation34]; bamlanivimab received EUA within 9 months of its initial discovery [Citation35], a considerably shorter timeline than for de novo small molecule antivirals [Citation36–38].

MAbs also have a very long duration of action, with half-lives of weeks [Citation15,Citation27]. Paxlovid and Remdesivir have half-lives of 6 and 0.89 hours, respectively [Citation39,Citation40]. In patients who are immunocompromised, the longer duration of action of mAbs may provide greater protection against severe COVID-19 over time. This is particularly important in comparison to Remdesivir which is also an IV infusion.

Small molecule antivirals are more likely than mAbs to have medication interactions and unintentional side effects due to off target binding and drug elimination. For example, Paxlovid is a strong CYP3A inhibitor, and is contraindicated for people taking medications that are either cleared by CYP3A, or strong CYP3A inducers that could reduce the plasma concentrations of Paxlovid [Citation41,Citation42]. If Paxlovid is being considered for use in high-risk pediatric populations, drug interactions for Paxlovid become an important consideration.

5. Indications for Covid-19 treatment in pediatric patients

The FDA has authorized mAb therapy for adolescents at high risk of severe COVID-19 despite limited data to support the use of mAbs in pediatric populations [Citation18,Citation43,Citation44]. Although the safety profile of these agents in adult studies is deemed acceptable, the safety, efficacy, and pharmacokinetics of mAbs have not been studied systematically in pediatric groups [Citation45,Citation46]. Consequently, the American Academy of Pediatrics and a separate panel of pediatric experts has recommended that, for adolescents at moderate and high risk for severe COVID-19, an individual risk/benefit assessment should be performed when considering the use of mAbs [Citation45,Citation46]. The panel did not recommend routine mAb treatment for lower risk patients [Citation46].

In general, children with COVID-19 are at low risk for hospitalization and progression to severe disease [Citation46,Citation47]. Conditions like obesity, medical complexity, and severe immunocompromising conditions may most strongly predispose children to severe COVID-19 [Citation46]. Immunocompromising conditions that place children at probable high risk include severe combined immunodeficiency (SCID) phenotypes, B-cell deficiencies, bone marrow transplant, and immune suppression for organ transplant [Citation48–55].

In rare cases, children with SARS-CoV-2 infection can develop Multisystem Inflammatory Syndrome in Children (MIS-C), a postinfectious complication of COVID-19 characterized by a severe shock-like illness [Citation56–58]. Common clinical features include fever, and gastrointestinal, cardiorespiratory, and neurocognitive symptoms [Citation59]. However, the disease presentation can have a wide spectrum and the risk factors for this condition are poorly understood [Citation60,Citation61].

Another complication of SARS-CoV-2 infection in children is ‘post-COVID-19 condition’ or ‘long-COVID.’ This is a broad range of physical and mental health symptoms that persist at least 4 weeks after the onset of symptoms [Citation62]. The prevalence of long-COVID in children and adolescents is 25% in one study; the most prevalent clinical manifestations are mood symptoms (16.5%), fatigue (9.7%), sleep disorders (8.4%), headache (7.8%), and respiratory symptoms (7.6%) [Citation63]. Whether antiviral treatments, including mAbs can modify the risk of Long-COVID in children remains unclear.

Severe COVID-19 is a public health concern for pediatric populations and disproportionately impacts Black, Hispanic, and American Indian/Alaska Native children in the United States [Citation64–66]. In one study, children hospitalized for COVID-19 are 2.28, 1.38, and 2.95 times more likely to be Black, Hispanic, and multi- racial/ethnic, respectively, than they are to be White [Citation67]. Children who died from COVID-19 are 2.96 and 3.33 times more likely to be Black or multiracial/ethnic than they are to be White [Citation67]. In another study, American Indian and Alaska Native patients were more likely to die of COVID-19 than all other races [Citation68]. MIS-C was also disproportionately high in Hispanic or Black children [Citation56,Citation69,Citation70]. The racial disparities are likely driven by social determinants of health such as socioeconomic status and access to health care, which may affect COVID-19 infection outcome directly or through elevated risk of high-risk conditions. Therefore, increasing treatment access to these disproportionately affected groups is especially important to ensure equitable care.

6. Deciding when to use monoclonal antibody therapy in children

Without sufficient data, assessing the risk-benefit of mAb treatment in pediatric patients is based largely on inferences from adult data, our understanding of pediatric physiology, and data on the natural history of SARS-CoV-2 infection in children. First, the administration of mAb therapy may reduce endogenous antibody production compared to placebo [Citation71,Citation72]. For severely immunocompromised individuals unable to produce an endogenous antibody-mediated immune response against SARS-CoV-2, this influence on endogenous immunity is of limited importance. However, the reduction in endogenous antibody production could impact risk for reinfection after recovery, and may influence vaccine-induced antibody response, particularly in patients receiving lower doses of vaccine [Citation73,Citation74]. Currently, we have inadequate evidence to determine whether this effect on antibody production has any clinically meaningful impact on long-term reinfection risk.

Another challenge regarding the use of mAb treatment for pediatric patients involves the ease of treatment access. MAb treatments authorized by the FDA are administered as intravenous or intramuscular injections by a health care provider, usually within 7 days of symptom onset [Citation15,Citation18–20]. The limited healthcare capacity for ambulatory infusions, space, and staffing may partially explain the underutilization of mAbs by high-risk populations [Citation75]. This barrier to treatment may be higher in children, as the limited available staff may also be less equipped to handle pediatric patients.

Compared to adults, pediatric patients are more likely to be asymptomatic or have mild symptoms when exposed to SARS-CoV-2. They are less likely to develop severe disease, be hospitalized, or die from COVID-19 [Citation76,Citation77]. Since children have less severe outcomes than adults with COVID-19 [Citation78], the likely high number needed to treat makes the rare side effects from treatment more important, and amplifies the direct (nursing time, travel costs) and indirect (patient time) costs associated with each beneficial clinical outcome.

7. Expert opinion

In patients with severely compromised immune systems or severe pulmonary disease at baseline, the benefits of antiviral treatment for COVID-19 likely outweigh the relatively small risks. However, for the great majority of pediatric patients, mAb treatment is likely not indicated. Whether the convenience of oral therapies such as Paxlovid outweigh the uncertain effects of the drug during child growth and development is also not clear is this time [Citation79]. The use of immunomodulators like convalescent plasma in pediatric populations also warrants more study [Citation80,Citation81].

7.1. Challenges to COVID-19 drug evaluation and approval

Numerous obstacles have influenced the rapid approval of mAb treatment for COVID-19. First, the SARS-CoV-2 virus has mutated over the course of the past year as it moves through hundreds of millions of people [Citation82]. This poses a challenge because a typical clinical trial timeline for a new mAb treatment takes years to conduct, and during that time, the virus will likely evade neutralization by the mAb being tested. This has happened already for mAbs that previously received EUAs and will likely continue to occur for mAbs developed in the future [Citation21,Citation25].

Since the mAbs are most effective against COVID-19 when given within about 7 days of symptom onset, from a logistical standpoint, a rapid pipeline from SARS-CoV-2 infection diagnosis to referral to treatment needs to be established. Many healthcare settings lack the infrastructure to facilitate this turnaround time, although solutions such as mobile infusion units, home health nursing, and outreach from government and community-based organizations, have emerged as cost-effect ways to address this [Citation83–86].

High recruitment for trials is another hurdle delaying new drug approvals. The risk of hospitalization for pediatric patients with SARS-CoV-2 infection is very low, and a majority of those hospitalized do not have severe disease [Citation76]. Consequently, the COVID-19 mAb treatment trials would need to have very large enrollment targets to demonstrate that the treatment prevents severe illness without leading to unacceptable toxicities. The need for large studies may be partially obviated by using surrogate endpoints such as viral load, though no secondary endpoints for hospitalization and death from SARS-CoV-2 have been definitively established, particularly in pediatric patients.

Finally, challenges persist regarding the use of placebo controls in mAb trials. Since bebtelovimab’s EUA, testing new mAbs against placebo is not ethically acceptable. COVID-19 treatments under EUA have become frequently used in clinical settings, and many participants and enrolling physicians are likely to see placebo controls as inferior to the EUA treatments, as most of the medical community currently does. If new mAb treatments were compared against existing ones to demonstrate superiority or lack of inferiority, this would require higher recruitment numbers. On the other hand, if new mAb trial drugs were compared to placebo, prospective participants may choose not to opt in, if they are aware that a potentially effective treatment already exists under EUA. Indeed, we observed this trend first-hand participating in mAb phase 3 testing after the initial EUAs were granted based on phase 2 data [Citation18]. In this case, many patients who were interested in enrolling in a trial for a new mAb were patients who did not meet the criteria to take a mAb with EUA but met inclusion criteria to enroll in the new mAb trial, or who otherwise did not have access to the EUA medication for logistical reasons. Both reasons introduce important biases into the final study data.

7.2. Conclusions

Vaccines are still the most effective strategy to stop and prevent the spread of COVID-19. However, in cases where vaccines are unable to prevent breakthrough infection, more treatments are needed. MAbs are an emerging therapy for COVID-19, however we have limited evidence for their safety and efficacy in children, and many have waning efficacy in the face of new variants. Small molecule antiviral therapies are another potential treatment for COVID-19 in children in an outpatient setting, though neither mAb nor small molecule antiviral treatments have significant supporting evidence in children. In rare circumstances, such as profound immunodeficiency, mAb treatments may still have an important role in treating children infected with SARS-CoV-2, though developing a strong evidence base for these decisions will be challenging if not impractical. Ultimately, these decisions are likely to be made at the level of individual cases using expert opinion as the primary guiding principle.

Article highlights

  • Anti-SARS-CoV-2 monoclonal antibodies (mAbs) have emerged as a potential treatment option to prevent hospitalization and death related to COVID-19.

  • Limited evidence exists for the use of monoclonal antibodies to treat COVID-19 in pediatric populations.

  • Small molecule antivirals and mAbs both show strong efficacy against COVID-19, but with different benefits and limitations.

  • In patients with severely compromised immune systems or severe pulmonary disease, the benefits of mAb treatment for COVID-19 likely outweigh the relatively small risks. However, for the great majority of pediatric patients, mAb treatment is likely not indicated.

  • Numerous obstacles have influenced the approval of mAb treatment for COVID-19, thus developing a strong evidence base for use of antivirals in children will be challenging if not impractical.

Declaration of interest

M Dougan has research funding from Eli Lilly; he has received consulting fees from Tillotts Pharma, ORIC Pharmaceuticals, Partner Therapeutics, SQZ Biotech, AzurRx, Eli Lilly, Mallinckrodt Pharmaceuticals, Aditum, and Moderna; he is a member of the Scientific Advisory Board for Neoleukin Therapeutics. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Acknowledgments

K Ling and M Dougan would like to thank Keri Sullivan for the thoughtful insights and revision of the manuscript.

Additional information

Funding

This paper was not funded.

References

  • Omer SB, Malani PN. Booster vaccination to prevent COVID-19 in the era of omicron: an effective part of a layered public health approach. JAMA. 2022;327(7):628–629. PubMed PMID: 35061011.
  • Chenchula S, Karunakaran P, Sharma S, et al.Current evidence on efficacy of COVID-19 booster dose vaccination against the Omicron variant: a systematic review.J Med Virol.2022;94(7):2969–2976. Epub 20220314. PubMed PMID: 35246846; PubMed Central PMCID: PMC9088621
  • Murdaca G, Noberasco G, Olobardi D, et al. Current take on systemic sclerosis patients’ vaccination recommendations. Vaccines (Basel). 2021;9(12). Epub 20211202. PubMed PMID: 34960174; PubMed Central PMCID: PMC8708328. doi: 10.3390/vaccines9121426.
  • Sadoff J, Gray G, Vandebosch A, et al. Safety and efficacy of single-dose Ad26.COV2.S vaccine against Covid-19. N Engl J Med. 2021;384(23):2187–2201. Epub 20210421. PubMed PMID: 33882225; PubMed Central PMCID: PMC8220996
  • Heath PT, Galiza EP, Baxter DN, et al. Safety and efficacy of NVX-CoV2373 Covid-19 vaccine. N Engl J Med. 2021;385(13):1172–1183. Epub 20210630. PubMed PMID: 34192426; PubMed Central PMCID: PMC8262625
  • Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020;383(27):2603–2615. Epub 20201210. PubMed PMID: 33301246; PubMed Central PMCID: PMC7745181
  • Frenck RW, Klein NP, Kitchin N, et al. Safety, immunogenicity, and efficacy of the BNT162b2 Covid-19 vaccine in adolescents. N Engl J Med. 2021;385(3):239–250. Epub 20210527. PubMed PMID: 34043894; PubMed Central PMCID: PMC8174030
  • Fowlkes AL, Yoon SK, Lutrick K, et al. Effectiveness of 2-Dose BNT162b2 (Pfizer BioNTech) mRNA vaccine in preventing SARS-CoV-2 infection among children aged 5-11 years and adolescents aged 12-15 years - PROTECT Cohort, July 2021-February 2022. MMWR Morb Mortal Wkly Rep. 2022;71(11):422–428. Epub 20220318. PubMed PMID: 35298453; PubMed Central PMCID: PMC8942308
  • Baden LR, El Sahly HM, Essink B, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2021;384(5):403–416. Epub 20201230. PubMed PMID: 33378609; PubMed Central PMCID: PMC7787219
  • Ali K, Berman G, Zhou H, et al. Evaluation of mRNA-1273 SARS-CoV-2 vaccine in adolescents. N Engl J Med. 2021;385(24):2241–2251. Epub 20210811. PubMed PMID: 34379915; PubMed Central PMCID: PMC8385554
  • Andrews N, Stowe J, Kirsebom F, et al. Covid-19 vaccine effectiveness against the omicron (B.1.1.529) variant. N Engl J Med. 2022;386(16):1532–1546. Epub 20220302. PubMed PMID: 35249272; PubMed Central PMCID: PMC8908811
  • Garcia-Beltran WF, St Denis KJ, Hoelzemer A, et al. mRNA-based COVID-19 vaccine boosters induce neutralizing immunity against SARS-CoV-2 Omicron variant. Cell. 2022;185(3):457–66.e4. Epub 20220106. PubMed PMID: 34995482; PubMed Central PMCID: PMC8733787
  • Marston HD, Paules CI, Fauci AS. Monoclonal antibodies for emerging infectious diseases - borrowing from history. N Engl J Med.2018;378(16):1469–1472. Epub 20180307. PubMed PMID: 29513615
  • Zhou D, Zhou R, Chen Z. Human neutralizing antibodies for SARS-CoV-2 prevention and immunotherapy. Immunother Adv. 2022;2(1):ltab027. Epub 20211230. PubMed PMID: 35915816; PubMed Central PMCID: PMC8755319
  • Dougan M, Azizad M, Chen P, et al. Bebtelovimab, alone or together with bamlanivimab and etesevimab, as a broadly neutralizing monoclonal antibody treatment for mild to moderate, ambulatory COVID-19. medRxiv. 2022:2022 March 10. 22272100. doi: 10.1101/2022.03.10.22272100.
  • Westendorf K, Žentelis S, Wang L, et al. LY-CoV1404 (bebtelovimab) potently neutralizes SARS-CoV-2 variants. Cell Rep. 2022;39(7):110812. Epub 20220425. PubMed PMID: 35568025; PubMed Central PMCID: PMC9035363
  • Gottlieb RL, Nirula A, Chen P, et al. Effect of bamlanivimab as monotherapy or in combination with etesevimab on viral load in patients with mild to moderate COVID-19: a randomized clinical trial. JAMA. 2021;325(7):632–644. PubMed PMID: 33475701; PubMed Central PMCID: PMC7821080.
  • Dougan M, Nirula A, Azizad M, et al. Bamlanivimab plus etesevimab in mild or moderate Covid-19. N Engl J Med. 2021;385(15):1382–1392. Epub 20210714. PubMed PMID: 34260849; PubMed Central PMCID: PMC8314785
  • Weinreich DM, Sivapalasingam S, Norton T, et al. REGN-COV2, a neutralizing antibody cocktail, in outpatients with Covid-19. N Engl J Med. 2021;384(3):238–251. Epub 20201217. PubMed PMID: 33332778; PubMed Central PMCID: PMC7781102
  • Gupta A, Gonzalez-Rojas Y, Juarez E, et al. Early treatment for Covid-19 with SARS-CoV-2 neutralizing antibody sotrovimab. N Engl J Med. 2021;385(21):1941–1950. Epub 20211027. PubMed PMID: 34706189
  • Takashita E, Kinoshita N, Yamayoshi S, et al. Efficacy of antibodies and antiviral drugs against Covid-19 omicron variant. N Engl J Med. 2022;386(10):995–998. Epub 20220126. PubMed PMID: 35081300; PubMed Central PMCID: PMC8809508
  • Tao K, Tzou PL, Kosakovsky Pond SL, et al. Susceptibility of SARS-CoV-2 omicron variants to therapeutic monoclonal antibodies: systematic review and meta-analysis. Microbiol Spectr. 2022;10(4):e0092622. Epub 20220614. PubMed PMID: 35700134.
  • Gupta A, Gonzalez-Rojas Y, Juarez E, et al. Effect of sotrovimab on hospitalization or death among high-risk patients with mild to moderate COVID-19: a randomized clinical trial. JAMA. 2022;327(13):1236–1246.
  • Weinreich DM, Sivapalasingam S, Norton T, et al. REGEN-COV antibody combination and outcomes in outpatients with Covid-19. N Engl J Med. 2021;385(23):e81. Epub 20210929. PubMed PMID: 34587383; PubMed Central PMCID: PMC8522800
  • Takashita E, Yamayoshi S, Simon V, et al. Efficacy of antibodies and antiviral drugs against omicron BA.2.12.1, BA.4, and BA.5 subvariants. N Engl J Med. 2022;387(5):468–470. Epub 20220720. PubMed PMID: 35857646; PubMed Central PMCID: PMC9342381 .
  • Iketani S, Liu L, Guo Y, et al. Antibody evasion properties of SARS-CoV-2 Omicron sublineages. Nature. 2022;604(7906):553–556. Epub 20220303. PubMed PMID: 35240676; PubMed Central PMCID: PMC9021018
  • Levin MJ, Ustianowski A, De Wit S, et al. Intramuscular AZD7442 (Tixagevimab-Cilgavimab) for prevention of Covid-19. N Engl J Med. 2022;386(23):2188–2200. Epub 20220420. PubMed PMID: 35443106; PubMed Central PMCID: PMC9069994
  • Wang M, Cao R, Zhang L, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30(3):269–271. Epub 20200204. PubMed PMID: 32020029; PubMed Central PMCID: PMC7054408
  • Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the treatment of Covid-19 - final report. N Engl J Med. 2020;383(19):1813–1826. Epub 20201008. PubMed PMID: 32445440; PubMed Central PMCID: PMC7262788
  • Goldman DL, Aldrich ML, Hagmann SHF, et al. Compassionate use of remdesivir in children with severe COVID-19. Pediatrics. 2021;147(5). PubMed PMID: 33883243. DOI:10.1542/peds.2020-047803.
  • Hammond J, Leister-Tebbe H, Gardner A, et al. Oral nirmatrelvir for high-risk, nonhospitalized adults with Covid-19. N Engl J Med. 2022;386(15):1397–1408. Epub 20220216. PubMed PMID: 35172054; PubMed Central PMCID: PMC8908851
  • Jayk Bernal A, da Silva Mm G, Musungaie DB, et al. Molnupiravir for oral treatment of Covid-19 in nonhospitalized patients. N Engl J Med. 2022;386(6):509–520. Epub 20211216. PubMed PMID: 34914868; PubMed Central PMCID: PMC8693688
  • Vangeel L, Chiu W, De Jonghe S, et al. Remdesivir, molnupiravir and nirmatrelvir remain active against SARS-CoV-2 Omicron and other variants of concern. Antiviral Res. 2022;198:105252. Epub 20220124. PubMed PMID: 35085683; PubMed Central PMCID: PMC8785409.
  • Smith K, Garman L, Wrammert J, et al. Rapid generation of fully human monoclonal antibodies specific to a vaccinating antigen. Nat Protoc. 2009;4(3):372–384. PubMed PMID: 19247287; PubMed Central PMCID: PMC2750034.
  • Chen P, Datta G, Grace Li Y, et al. First-in-human study of bamlanivimab in a randomized trial of hospitalized patients with COVID-19. Clin Pharmacol Ther. 2021;110(6):1467–1477. Epub 20211016. PubMed PMID: 34455583; PubMed Central PMCID: PMC8653186
  • Riva L, Yuan S, Yin X, et al. Discovery of SARS-CoV-2 antiviral drugs through large-scale compound repurposing. Nature. 2020;586(7827):113–119. Epub 20200724. PubMed PMID: 32707573; PubMed Central PMCID: PMC7603405
  • Bolken TC, Hruby DE. Discovery and development of antiviral drugs for biodefense: experience of a small biotechnology company. Antiviral Res.2008;77(1):1–5; Epub 20070815. PubMed PMID: 17765333; PubMed Central PMCID: PMC2972676;
  • Ward DJ, Hammond E, Linden-Phillips L, et al.Trends in clinical development timeframes for antiviral drugs launched in the UK, 1981-2014: a retrospective observational study.BMJ Open.2015;5(11):e009333. Epub 20151116. PubMed PMID: 26576812; PubMed Central PMCID: PMC4654359
  • Lamb YN. Nirmatrelvir plus ritonavir: first approval. Drugs. 2022;82(5):585–591. PubMed PMID: 35305258; PubMed Central PMCID: PMC8933659.
  • Tempestilli M, Caputi P, Avataneo V, et al. Pharmacokinetics of remdesivir and GS-441524 in two critically ill patients who recovered from COVID-19. J Antimicrob Chemother. 2020;75(10):2977–2980. PubMed PMID: 32607555; PubMed Central PMCID: PMC7337789.
  • Eng H, Dantonio AL, Kadar EP, et al. Disposition of nirmatrelvir, an orally bioavailable inhibitor of SARS-CoV-2 3C-Like Protease, across animals and humans. Drug Metab Dispos. 2022;50(5):576–590. Epub 20220213. PubMed PMID: 35153195
  • Hsu A, Granneman GR, Bertz RJR. Clinical pharmacokinetics and interactions with other anti-HIV agents. Clin Pharmacokinet. 1998;35(4):275–291. PubMed PMID: 9812178.
  • Mak G, Dassner AM, Hammer BM, et al. Safety and tolerability of monoclonal antibody therapies for treatment of COVID-19 in pediatric patients. Pediatr Infect Dis J. 2021;40(12):e507–e9. PubMed PMID: 34260492; PubMed Central PMCID: PMC8575082.
  • Romani L, Calò Carducci FI, Chiurchiù S, et al. Safety of monoclonal antibodies in children affected by SARS-CoV-2 infection. Children (Basel). 2022;9(3). Epub 20220307. PubMed PMID: 35327741; PubMed Central PMCID: PMC8947030. doi: 10.3390/children9030369.
  • Wolf J, Abzug MJ, Wattier RL, et al. Initial guidance on use of monoclonal antibody therapy for treatment of coronavirus disease 2019 in children and adolescents. J Pediatric Infect Dis Soc. 2021;10(5):629–634. PubMed PMID: 33388760; PubMed Central PMCID: PMC7799019.
  • Wolf J, Abzug MJ, Anosike BI, et al.Updated guidance on use and prioritization of monoclonal antibody therapy for treatment of COVID-19 in adolescents. J Pediatric Infect Dis Soc. 2022;11(5):177–185. PubMed PMID: 35107571; PubMed Central PMCID: PMC8903349.
  • Bailey LC, Razzaghi H, Burrows EK, et al. Assessment of 135 794 pediatric patients tested for severe acute respiratory syndrome coronavirus 2 across the United States. JAMA Pediatr. 2021;175(2):176–184.
  • Delavari S, Abolhassani H, Abolnezhadian F, et al. Impact of SARS-CoV-2 pandemic on patients with primary immunodeficiency. J Clin Immunol. 2021;41(2):345–355. Epub 20201201. PubMed PMID: 33263173; PubMed Central PMCID: PMC7707812
  • London J, Boutboul D, Lacombe K, et al. Severe COVID-19 in patients with B cell alymphocytosis and response to convalescent plasma therapy. J Clin Immunol. 2021;41(2):356–361. Epub 20201120. PubMed PMID: 33219491; PubMed Central PMCID: PMC7678568
  • Hensley MK, Bain WG, Jacobs J, et al. Intractable coronavirus disease 2019 (COVID-19) and prolonged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication in a chimeric antigen receptor-modified T-cell therapy recipient: a case study. Clin Infect Dis. 2021;73(3):e815–e21. PubMed PMID: 33507235; PubMed Central PMCID: PMC7929077.
  • Truong TT, Ryutov A, Pandey U, et al. Increased viral variants in children and young adults with impaired humoral immunity and persistent SARS-CoV-2 infection: a consecutive case series. EBioMedicine. 2021;67:103355. Epub 20210426. PubMed PMID: 33915337; PubMed Central PMCID: PMC8072072.
  • Sharma A, Bhatt NS, St Martin A, et al. Clinical characteristics and outcomes of COVID-19 in haematopoietic stem-cell transplantation recipients: an observational cohort study. Lancet Haematol. 2021;8(3):e185–e93. Epub 20210119. PubMed PMID: 33482113; PubMed Central PMCID: PMC7816949
  • Goss MB, Galván NTN, Ruan W, et al. The pediatric solid organ transplant experience with COVID-19: an initial multi-center, multi-organ case series. Pediatr Transplant. 2021;25(3):e13868. Epub 20201109. PubMed PMID: 32949098; PubMed Central PMCID: PMC7537006
  • Hippisley-Cox J, Coupland CA, Mehta N, et al. Risk prediction of covid-19 related death and hospital admission in adults after covid-19 vaccination: national prospective cohort study. BMJ. 2021;374(2244). Epub 20210917. PubMed PMID: 34535466; PubMed Central PMCID: PMC8446717. doi: 10.1136/bmj.n2244.
  • Murdaca G, Greco M, Tonacci A, et al. IL-33/IL-31 axis in immune-mediated and allergic diseases. Int J Mol Sci. 2019;20(23):5856. Epub 20191122. PubMed PMID: 31766607; PubMed Central PMCID: PMC6929191
  • Feldstein LR, Tenforde MW, Friedman KG, et al. Characteristics and outcomes of US children and adolescents with multisystem inflammatory syndrome in children (MIS-C) compared with severe acute COVID-19. JAMA. 2021;325(11):1074–1087. PubMed PMID: 33625505; PubMed Central PMCID: PMC7905703.
  • Nakra NA, Blumberg DA, Herrera-Guerra A, et al. Multi-system inflammatory syndrome in children (MIS-C) following SARS-CoV-2 infection: review of clinical presentation, hypothetical pathogenesis, and proposed management. Children (Basel). 2020;7(7). Epub 20200701. PubMed PMID: 32630212; PubMed Central PMCID: PMC7401880. doi:10.3390/children7070069.
  • Radia T, Williams N, Agrawal P, et al. Multi-system inflammatory syndrome in children & adolescents (MIS-C): a systematic review of clinical features and presentation. Paediatr Respir Rev. 2021;38:51–57. Epub 20200811. PubMed PMID: 32891582; PubMed Central PMCID: PMC7417920.
  • Whittaker E, Bamford A, Kenny J, et al. Clinical characteristics of 58 children with a pediatric inflammatory multisystem syndrome temporally associated with SARS-CoV-2. JAMA. 2020;324(3):259–269. PubMed PMID: 32511692; PubMed Central PMCID: PMC7281356.
  • Abrams JY, Oster ME, Godfred-Cato SE, et al. Factors linked to severe outcomes in multisystem inflammatory syndrome in children (MIS-C) in the USA: a retrospective surveillance study. Lancet Child Adolesc Health. 2021;5(5):323–331. Epub 20210310. PubMed PMID: 33711293; PubMed Central PMCID: PMC7943393
  • Esposito S, Caramelli F, Principi N. What are the risk factors for admission to the pediatric intensive unit among pediatric patients with COVID-19? Ital J Pediatr.2021;47(1):103. Epub 20210503. PubMed PMID: 33941228; PubMed Central PMCID: PMC8090919
  • Nalbandian A, Sehgal K, Gupta A, et al. Post-acute COVID-19 syndrome. Nat Med. 2021;27(4):601–615. Epub 20210322. PubMed PMID: 33753937; PubMed Central PMCID: PMC8893149
  • Lopez-Leon S, Wegman-Ostrosky T, Ayuzo Del Valle NC, et al. Long-COVID in children and adolescents: a systematic review and meta-analyses. Sci Rep. 2022;12(1):9950. Epub 20220623. PubMed PMID: 35739136; PubMed Central PMCID: PMC9226045
  • White A, Liburd LC, Coronado F. Addressing racial and ethnic disparities in COVID-19 among school-aged children: are we doing enough? Prev Chronic Dis. 2021;18:E55. Epub 20210603. PubMed PMID: 34081577; PubMed Central PMCID: PMC8220967.
  • Gold JAW, Rossen LM, Ahmad FB, et al. Race, ethnicity, and age trends in persons who died from COVID-19 - United States, May-August 2020. MMWR Morb Mortal Wkly Rep. 2020;69(42):1517–1521. Epub 20201023. PubMed PMID: 33090984; PubMed Central PMCID: PMC7583501
  • Kim L, Whitaker M, O’Halloran A, et al. Hospitalization rates and characteristics of children aged <18 years hospitalized with laboratory-confirmed COVID-19 - COVID-NET, 14 states, March 1-July 25, 2020. MMWR Morb Mortal Wkly Rep. 2020;69(32):1081–1088. Epub 20200814. PubMed PMID: 32790664; PubMed Central PMCID: PMC7440125
  • Moreira A, Chorath K, Rajasekaran K, et al.Demographic predictors of hospitalization and mortality in US children with COVID-19.Eur J Pediatr.2021;180(5):1659–1663. Epub 20210120. PubMed PMID: 33474580; PubMed Central PMCID: PMC7817069
  • Musshafen LA, El-Sadek L, Lirette ST, et al.In-hospital mortality disparities among American Indian and Alaska native, black, and white patients with COVID-19.JAMA Network Open.2022;5(3):e224822. Epub 20220301. PubMed PMID: 35353166; PubMed Central PMCID: PMC8968465
  • Abrams JY, Godfred-Cato SE, Oster ME, et al. Multisystem inflammatory syndrome in children associated with severe acute respiratory syndrome coronavirus 2: a systematic review. J Pediatr. 2020;226:45–54.e1. Epub 20200805. PubMed PMID: 32768466; PubMed Central PMCID: PMC7403869.
  • Payne AB, Gilani Z, Godfred-Cato S, et al. Incidence of multisystem inflammatory syndrome in children among US persons infected with SARS-CoV-2. JAMA Network Open. 2021;4(6):e2116420. Epub 20210601. PubMed PMID: 34110391; PubMed Central PMCID: PMC8193431
  • Zhang L, Poorbaugh J, Dougan M, et al. Endogenous antibody responses to SARS-CoV-2 in patients with mild or moderate COVID-19 who received bamlanivimab alone or bamlanivimab and etesevimab together. Front Immunol. 2021;12:790469. Epub 20211209. PubMed PMID: 34956222; PubMed Central PMCID: PMC8697104.
  • Kim PS, Dimcheff DE, Siler A, et al. Effect of monoclonal antibody therapy on the endogenous SARS-CoV-2 antibody response. Clin Immunol. 2022;236:108959. Epub 20220224. PubMed PMID: 35218964; PubMed Central PMCID: PMC8866167
  • Lucas C, Klein J, Sundaram ME, et al. Delayed production of neutralizing antibodies correlates with fatal COVID-19. Nat Med. 2021;27(7):1178–1186. Epub 20210505. PubMed PMID: 33953384; PubMed Central PMCID: PMC8785364
  • Khoury DS, Cromer D, Reynaldi A, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med. 2021;27(7):1205–1211. Epub 20210517. PubMed PMID: 34002089
  • Wu EL, Kumar RN, Moore WJ, et al. Disparities in COVID-19 monoclonal antibody delivery: a retrospective cohort study. J Gen Intern Med. 2022;37(10):2505–2513. Epub 20220425. PubMed PMID: 35469360; PubMed Central PMCID: PMC9037582
  • Zsigmond B, Breathnach AS, Mensah A, et al.Very low rates of severe COVID-19 in children hospitalised with confirmed SARS-CoV-2 infection in London, England”.J Infect.2022;85(1):90–122. Epub 20220410. PubMed PMID: 35417737.
  • Smith C, Odd D, Harwood R, et al. Deaths in children and young people in England after SARS-CoV-2 infection during the first pandemic year. Nat Med. 2022;28(1):185–192. Epub 20211111. PubMed PMID: 34764489
  • Zimmermann P, Curtis N. Why is COVID-19 less severe in children? A review of the proposed mechanisms underlying the age-related difference in severity of SARS-CoV-2 infections. Arch Dis Child. 2020; Epub 20201201. PubMed PMID: 33262177. doi: 10.1136/archdischild-2020-320338.
  • Yan G, Zhou J, Zhu H, et al. The feasibility, safety, and efficacy of Paxlovid treatment in SARS-CoV-2-infected children aged 6-14 years: a cohort study. Ann Transl Med. 2022;10(11):619. PubMed PMID: 35813342; PubMed Central PMCID: PMC9263777.
  • Dulek DE, Fuhlbrigge RC, Tribble AC, et al. Multidisciplinary guidance regarding the use of immunomodulatory therapies for acute coronavirus disease 2019 in pediatric patients. J Pediatric Infect Dis Soc. 2020;9(6):716–737. PubMed PMID: 32808988; PubMed Central PMCID: PMC7454742.
  • Zaffanello M, Piacentini G, Nosetti L, et al.The use of convalescent plasma for pediatric patients with SARS-CoV-2: a systematic literature review.Transfus Apher Sci.2021;60(2):103043. Epub 20201223. PubMed PMID: 33388249; PubMed Central PMCID: PMC7834628
  • Morales AC, Rice AM, Ho AT, et al. Causes and Consequences of Purifying Selection on SARS-CoV-2. Genome Biol Evol. 2021;13(10). PubMed PMID: 34427640; PubMed Central PMCID: PMC8504154. DOI:10.1093/gbe/evab196.
  • Tulledge-Scheitel S, Bell SJ, Larsen JJ, et al. A mobile unit overcomes the challenges to monoclonal antibody infusion for COVID-19 in skilled care facilities. J Am Geriatr Soc. 2021;69(4):868–873. Epub 20210305. PubMed PMID: 33619724; PubMed Central PMCID: PMC8013486
  • Malani AN, LaVasseur B, Fair J, et al. Administration of monoclonal antibody for COVID-19 in patient homes. JAMA Network Open. 2021;4(10):e2129388. Epub 20211001. PubMed PMID: 34648014; PubMed Central PMCID: PMC8517740
  • Walker VL, Meyer A, Paauw D, et al.Health system success in delivering bamlanivimab within post-acute and long-term care facilities.J Am Med Dir Assoc.2022;23(1):141–145. Epub 20211208. PubMed PMID: 34890559; PubMed Central PMCID: PMC8651463;
  • Razonable RR, Aloia NCE, Anderson RJ, et al. A framework for outpatient infusion of antispike monoclonal antibodies to high-risk patients with mild-to-moderate coronavirus disease-19: the mayo clinic model. Mayo Clin Proc. 2021;96(5):141–145. Epub 20210309. PubMed PMID: 33958056; PubMed Central PMCID: PMC7942148