Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Apr 6, 2023; 11(10): 2168-2180
Published online Apr 6, 2023. doi: 10.12998/wjcc.v11.i10.2168
Clinical application of SARS-CoV-2 antibody detection and monoclonal antibody therapies against COVID-19
Jin Sun, Bei-Bei Qu, Medical Innovation Research Office, Sinopharm Gezhouba Central Hospital, Third Clinical Medical College of Three Gorges University, Yichang 443002, Hubei Province, China
Zhen-Dong Yang, Department of Respiratory, Beijing Jindu Children Hospital, Beijing 102208, China
Zhen-Dong Yang, Innovative Medicine Working Committee of the Chinese Society of Water Resources and Electric Power Medical Science and Technology, Beijing 100053, China
Zhen-Dong Yang, Xiong Xie, Department of Pediatrics, Sinopharm Gezhouba Central Hospital, Third Clinical Medical College of Three Gorges University, Yichang 443002, Hubei Province, China
Li Li, Department of Intensive Care, First Clinical Medical College of Three Gorges University, Yichang 443000, Hubei Province, China
Hua-Song Zeng, Department of Allergy Immunology and Rheumatology, Guangzhou Children's Hospital, Women's and Children's Medical Center Affiliated with Guangzhou Medical University, Guangzhou 510000, Guangdong Province, China
Bo Gong, Central Laboratory, Shanghai Changning District Maternal and Child Health, Maternal and Child Health Hospital Affiliated with Shanghai East China Normal University, Shanghai 210000, China
Jian-Qiang Xu, Department of Respiratory and Critical Care Medicine, Sinopharm Gezhouba Central Hospital, Third Clinical Medical College of Three Gorges University, Yichang 443002, Hubei Province, China
Ji-Hong Wu, School of Clinical Medicine, Beijing Tsinghua Chang Gung Hospital Affiliated to Tsinghua University, Beijing 102218, China
Guo-Wei Song, Department of Emergency, Children's Hospital Affiliated with Beijing Capital Institute of Pediatrics, Beijing 100020, China
ORCID number: Jin Sun (0000-0002-6308-1958); Zhen-Dong Yang (0000-0003-0719-7400); Xiong Xie (0000-0003-4990-6703); Li Li (0000-0003-0928-1796); Hua-Song Zeng (0000-0002-7166-0941); Bo Gong (0000-0002-4793-7699); Jian-Qiang Xu (0000-0002-6990-3289); Ji-Hong Wu (0000-0002-6232-0187); Bei-Bei Qu (0000-0003-2113-8968); Guo-Wei Song (0000-0002-9356-9145).
Author contributions: Yang ZD and Sun J contributed to conception and design, these authors have contributed equally to this work and share first authorship; Yang ZD, Qu BB, Song GW, Xu JQ and Gong B contributed to administrative support; Song GW and Li L contributed to provision of study materials; Yang ZD, Song GW , Xie X, Li L, Zeng HS and Wu JH contributed to collection and assembly of data; Yang ZD, Sun J, Zeng HS, Gong B, Xu JQ, Qu BB and Xie X contributed to data analysis and interpretation; All authors contributed to manuscript revision and approved the submitted version.
Conflict-of-interest statement: All the authors report having no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Zhen-Dong Yang, Chief Physician, Professor, Department of Respiratory, Beijing Jindu Children Hospital, No. 308 Huilongguan East Street, Changping district, Beijing 102208, China. yzd886@sina.com
Received: December 7, 2022
Peer-review started: December 7, 2022
First decision: January 5, 2023
Revised: January 17, 2023
Accepted: March 10, 2023
Article in press: March 10, 2023
Published online: April 6, 2023

Abstract

The purpose of this study was to investigate the clinical application of severe acute respiratory distress syndrome coronavirus-2 (SARS-CoV-2) specific antibody detection and anti-SARS-CoV-2 specific monoclonal antibodies (mAbs) in the treatment of coronavirus infectious disease 2019 (COVID-19). The dynamic changes of SARS-CoV-2 specific antibodies during COVID-19 were studied. Immunoglobulin M (IgM) appeared earlier and lasted for a short time, while immunoglobulin G (IgG) appeared later and lasted longer. IgM tests can be used for early diagnosis of COVID-19, and IgG tests can be used for late diagnosis of COVID-19 and identification of asymptomatic infected persons. The combination of antibody testing and nucleic acid testing, which complement each other, can improve the diagnosis rate of COVID-19. Monoclonal anti-SARS-CoV-2 specific antibodies can be used to treat hospitalized severe and critically ill patients and non-hospitalized mild to moderate COVID-19 patients. COVID-19 convalescent plasma, highly concentrated immunoglobulin, and anti-SARS-CoV-2 specific mAbs are examples of anti-SARS-CoV-2 antibody products. Due to the continuous emergence of mutated strains of the novel coronavirus, especially omicron, its immune escape ability and infectivity are enhanced, making the effects of authorized products reduced or invalid. Therefore, the optimal application of anti-SARS-CoV-2 antibody products (especially anti-SARS-CoV-2 specific mAbs) is more effective in the treatment of COVID-19 and more conducive to patient recovery.

Key Words: SARS-CoV-2 antibody, Detection, COVID-19, Monoclonal antibody, Clinical application

Core Tip: Immunoglobulin M testing can be used for early diagnosis of coronavirus infectious disease 2019 (COVID-19). Immunoglobulin G testing can be used for the late diagnosis of COVID-19 and the identification of asymptomatic patients. The combination of antibody and nucleic acid testing has improved the diagnosis rate of COVID-19. The continuous emergence of mutated strains of the novel coronavirus, especially omicron, enhances its immune escape ability and infectivity, making the effects of authorized products reduced or invalid. The specific monoclonal antibodies against severe acute respiratory distress syndrome coronavirus-2 authorized by the United States Food and Drug Administration are more beneficial for the treatment of COVID-19 and patient recovery.



INTRODUCTION

Since the coronavirus infectious disease 2019 (COVID-19) pandemic began in 2019, it has had a devastating impact on communities across the globe. So far, severe acute respiratory distress syndrome coronavirus-2 (SARS-CoV-2) has mutated several times with the identification of the following variant strains: Alpha (B.1.1.7) was first discovered in the United Kingdom in late December 2020; Beta (B.1.351) was first reported in South Africa in December 2020; Gamma was first reported in Brazil in early January 2021; Delta (B.1.617.2) was first reported in India in December 2020; Omicron (B.1.1.529) was first reported in South Africa in November 2021 and quickly spread to countries around the world due to its increased infectivity. Omicron's spike protein has exhibited more than 30 changes that enhanced viral capacity for immune escape. Studies have shown that Omicron shows a 13-fold increase in viral infectivity, and is 2.8 times more infectious than the delta variant, and previously approved monoclonal antibodies (mAbs) against SARS-CoV-2 are less effective against this variant. Furthermore, vaccines against SARS-CoV-2 are less effective in prevention of Omicron infection, and treatment is more challenging[1]. For these reasons, Omicron has become a major variant of concern in many countries, and many mutants of this strain have been identified (e.g., BA.1, BA.2, BA.3, BA.4, BA.5, and XBB.1.5). The XBB1.5 mutant is becoming increasingly frequent in the United States, and is still defined as an Omicron lineage variant[2]. It remains to be seen whether this specific mutant strain will worsen the ongoing COVID-19 pandemic.

As of January 13, 2023, there have been over 660 million confirmed COVID-19 cases and over 6.7 million deaths reported to the World Health Organization (WHO)[3]. The timely and accurate diagnosis of COVID-19 is necessary for controlling the spread of the virus, initiating prompt treatment, and rehabilitating patients. The primary method for diagnosing COVID-19 is via detection of SARS-CoV-2 RNA using polymerase chain reaction (PCR). However, the accuracy of PCR analysis depends on many factors, such as the quality of the reagents, the quality and method of sample collection (oropharynx, nasopharynx, lower respiratory tract, and swabs), and the skill level of the operator. Changes in the above factors may lead to either false negative or false positive results. PCR has many additional limitations: long testing time (average 2–3 h); complex technology; easy contamination of samples; high cost; and requirement for a laboratory with strict biosafety certification and trained technicians. These issues pose serious challenges in screening for COVID-19, achieving early diagnosis, implementing isolation of infected patients, and starting treatment. As such, new diagnostic tools are urgently needed to help compensate for the shortcomings of PCR[4].

The detection of SARS-CoV-2 specific antibodies (SSAs) can aid in the diagnosis of COVID-19. Additionally, asymptomatic infected people and those with milder symptoms can also be identified, helping to calculate the population infection rate more accurately. Large scale serological maps of a population can also be used in epidemiological investigations to provide a better understanding of the pandemic status, help to control the spread of the virus, and aid in treating patients efficiently. Antibody detection and nucleic acid detection can complement each other and improve the rate of accurate COVID-19 diagnosis. Indeed, serological detection technology has been widely used in the clinic[4].

SARS-CoV-2 induces an excessive and prolonged cytokine/chemokine response, known as a cytokine storm, in some infected individuals, particularly those with severe COVID-19 disease. Cytokine storm can cause acute respiratory distress syndrome or multiple organ dysfunction, which can lead to deterioration or death. Timely control of early cytokine storm with administration of immunomodulators and cytokine antagonists can reduce inflammatory cell infiltration into the lung and limit the disease. Suppressing cytokine storm effectively is important to prevent deterioration in COVID-19 patients and key to preventing death associated with this disease[5].

mAbs are immunoglobulins that are produced from single-cell lineages and have a high affinity for their target cells. When used as antiviral therapy, neutralizing Abs (Nabs) aid in the development of passive antiviral immunity. The viral genome is susceptible to mutation, resulting in the emergence of viral escape variants that render the virus resistant to specific mAbs. Several mAbs were combined into cocktails to improve efficacy and overcome the phenomenon of viral escape. The United States Food and Drug Administration (FDA) has granted emergency use authorization (EUA) for 4 mAbs. As new SARS-CoV-2 variants emerge, the clinical efficacy of previously approved mAbs is being called into question, prompting clinicians and scientists to reconsider and pursue the development of new drugs to treat COVID-19. The FDA has also made numerous updates to previously authorized recommended drugs, rendering them unavailable or making conditional recommendations.

Convalescent plasma (CP), highly concentrated immunoglobulin (HIG), and anti-SSA (ASSA) products have been recommended or conditionally recommended for use in clinical research and healthcare facilities. For patients with mild to moderate COVID-19 who are at risk of progressing to critical illness, the FDA has approved the use of single or combined mAbs. Two mAbs can be combined in some cases to interfere with SARS-CoV-2 for post-exposure prophylaxis. Tocilizumab, an anti-interleukin-6 inhibitor, and baricitinib, a Janus kinase inhibitor, have received EUA from the FDA and are recommended for patients with severe COVID-19 who are receiving corticosteroids[6]. Tocilizumab reduces mortality in critically ill patients and also the number of days without need for artificial organ support; the recommended dose is 8 mg tocilizumab per kg body weight (up to 800 mg) in 1 h intravenous infusion, with a second dose repeated 12 to 24 h later if needed[7]. The combination of interleukin-6 blockers in particular may be more beneficial[8]. Baricitinib, an antiviral drug, is used to treat severe or critical COVID-19 and has been shown to reduce mortality, hospitalization, and the need for mechanical ventilation[9,10]. Corticosteroids are usually only used as a last resort in COVID-19 patients who are critically ill; routine use is generally not advised.

The purpose of this article is to discuss the clinical value of SSA detection technologies and ASSA in the treatment of COVID-19.

DYNAMIC CHANGES IN SARS-COV-2 SPECIC ANTIBODIES

Antibodies are reactive substances (immunoglobulins) produced by the immune system to combat invading pathogenic microorganisms and can bind to specific pathogens. SSAs are mainly composed of immunoglobulin M (IgM) and immunoglobulin G (IgG) Abs. IgM Abs are produced during the early stages of infection and can enhance the ability of the innate immune system to phagocytose, agglutinate, and kill pathogens. IgG is the main type of Ab in plasma (approximately 75%) and is involved in Ab-dependent cellular phagocytosis and complement-mediated cytotoxicity against bacterial and viral infections. Abs positivity is indirect evidence of infection caused by pathogenic microorganisms. Detecting the levels of IgM and IgG can also reveal the immune function of the body, which is an important factor contributing to public health and has great clinical significance.

After infection by a pathogen, certain steps associated with the production of Abs can be used to diagnose and treat disease. In a COVID-19 study involving 34 adults, Abs were assessed starting at 2 wk after disease onset. After 3 wk, the Ab tests of all patients were positive. The average levels of IgG and IgM were 112.40 AU/mL and 322.80 AU/mL (reference range < 10 AU/mL), respectively. After 4 wk, the level of IgM began to decline, and an average value of 147.92 AU/mL was recorded; the average level of IgG increased to 157.01 AU/mL. After 5 wk, the average level of IgM continued to fall, reaching 78.03 AU/mL, and 2 patients were negative for IgM; the IgG levels of all patients increased, reaching an average of 163.56 AU/mL. At the end of week 7, the study was terminated. Two patients were negative for IgM, and the average IgM level dropped to 21.83 AU/mL; all patients were positive for IgG, and their average titer increased to 167.16 AU/mL. A high level of IgM indicates that the infection is in the acute phase, and relatively high IgM levels lasting longer than 1 mo indicate a longer viral replication time. IgG Abs appear later than IgM Abs, persist longer, and are present at a higher level, indicating that the humoral immune response acts to defend the host against SARS-CoV-2 infection[11].

A study by Lee et al[12] showed that IgM was detected on the day 5 of infection, and the longest duration of detection was 42 d after the onset of disease. In another patient, IgG was also detected by day 5 after the onset of disease. In most cases, once positive, IgG remains positive. The duration of PCR positivity is related to the Abs response and clinical manifestations. In patients with detectable symptoms and IgM Abs positivity, nucleic acid-based tests can quickly become negative[12]. Another study analyzed 38 cases of COVID-19 and found the highest level of viral RNA in sputum samples (92.3%) in the early stage of the disease (7 d after disease onset); the next highest viral RNA level was detected in pharyngeal swabs (69.2%). In this study, Abs were detected in fewer positive cases (IgM 23.0% and IgG 53.8%). Abs titers increased to 50.0% (IgM) and 87.5% (IgG) after 8 d, and Abs detection was found to be easier than viral RNA detection. After 15 d, the detection rate of viral RNA was 52.2%, indicating that Abs detection could be useful for auxiliary diagnosis. For patients who have achieved clearance of the virus, PCR testing is not suitable. The initial immune response is marked by an increase in the IgM level and the detection of IgM indicates that the infection is recent; as such, IgM can be used for the early diagnosis of COVID-19. IgG is produced after IgM and persists longer; it is a diagnostic indicator of a secondary infection or previous exposure[13].

Timilsina et al[14], described an electrochemical device that can rapidly and quantitatively detect Abs against SARS-CoV-2 in clinic. This method was shown to be simple to use, economic, sensitive (100% sensitivity and specificity), and fast (10 min). Furthermore, it required blood samples of only 1.5 μL, and Abs could even be detected in dry blood spots. This test can be operated on-site and via remote application, and may have great promise in the future of COVID-19 testing[14].

The monumental achievement in creating an effective vaccine against SARS-CoV-2 presents a new challenge for Abs testing: can Abs produced by vaccination be distinguished from those produced by the real virus? How long do the Abs from the vaccine last? The answers to these questions are key to preparing for any future wave of the COVID-19 pandemic[15]. A schematic diagram of the dynamic changes in human antibody production after the initial infection (Figure 1).

Figure 1
Figure 1 Schematic diagram of the dynamic changes in antibody production after initial infection with severe acute respiratory virus-2. Due to differences in data from analytical studies, these data are considered approximations only.

Zamani et al[16] studied the kinetics of seroconversion in 118 hospitalized patients with COVID-19 (Table 1). The authors demonstrated the kinetics of seroconversion in COVID-19 patients, and showed that 83.5% of IgM and 36.8% of IgG patients who were originally positive were found to be negative within 3 mo after the onset of symptoms. These findings suggest that the retention time of anti-SARS-CoV-2 Abs could be used as a reference for SARS-CoV-2 prevention, control, and vaccine administration. No correlation was found between Abs titers and patient age at any time point. In addition, mean Abs levels at different times after the onset of clinical symptoms also did not exhibit a predilection for either sex.

Table 1 Seropositive rates of severe acute respiratory distress syndrome coronavirus-2-specific immunoglobulin G and immunoglobulin M at various time points after symptom onset.
Days after symptom onset
IgM positivity rate (%)
IgG positivity rate (%)
0–544/118 (37.3)71/118 (60.2)
6–1536/55 (65.5)44/55 (80.0)
16–2548/70 (68.6)56/70 (80.0)
26–3526/56 (46.4)2/56 (75.0)
36–9517/95 (17.9)56/95 (58.9)
ROLE OF SARS-COV-2 SPECIFIC ANTIBODIES IN COVID-19 SCREENING AND DIGNOSIS

Wu et al[17] performed COVID-19 screening on 1021 workers in the Recovery Work Group (RG) and 381 patients in the COVID-19 Non-Hospital Group (NHG), totaling 1402 subjects. Screening included nasopharyngeal swab nucleic acid detection, chest computed tomography (CT), and IgM and IgG Abs detection. In NHG, one case of nucleic acid was positive, as were IgM and IgG. Other patients' CT chest radiographs were normal, and the nucleic acid tests were negative. After being diagnosed with COVID-19, the positive patient was "cured." He was transferred to a hospital specializing in COVID-19 treatment after testing positive again. In RG, 98 (98/1021, 9.60%) patients were IgG positive, IgM negative, and nasopharyngeal swab PCR negative. In NHG, 39 cases (39/380, 10.3%) of nasopharyngeal swab samples were positive for IgG, negative for IgM, and negative by PCR. These findings suggest that 137 (98 + 39) patients had previously been infected with SARS-CoV-2. The IgG-positive patients had no suspicious symptoms and no history of COVID-19. None of these cases had previously been tested by PCR. In other words, the patients had recovered from asymptomatic infections discovered only through Abs screening[17].

To determine the diagnostic efficacy of IgM and IgG Abs in suspected cases, Abs tests were conducted in 52 patients whose symptoms or CT imaging characteristics met the COVID-19 criteria but who had multiple consecutive negative PCR results. IgG or IgM Abs were positive in 4 patients. Reasonable speculation led to a diagnosis of COVID-19. Similarly, to determine the diagnostic efficacy of IgG and IgM tests for asymptomatic infections, the team conducted Ab screening in 164 close contacts, 16 of whom were positive for IgM or IgG. Among the 148 patients excluded by PCR 1 mo before the screening process, 7 were also positive for the Abs. These individuals had never experienced any symptoms, indicating that they were asymptomatic carriers who could not have been diagnosed by nucleic acid detection. Thus, increasing the frequency of IgM and IgG Abs testing could help to confirm the diagnosis of suspected cases and screen for asymptomatic individuals[18].

A comprehensive study found that the earliest IgM Abs appear within 5–7 d after infection with SARS-CoV-2[13]. Abs testing can provide results within 15 min, and the technique is simple and convenient, which is helpful for the rapid screening of febrile patients. In infected individuals, during the incubation period of the virus before the development of the disease, IgM is not produced and cannot be detected. Hence, the detection of Abs alone is not suitable for early screening. Abs testing can only play an auxiliary diagnostic role; the maintenance time of IgM is short, and IgM disappears by the end of the infection. IgM positivity is the only basis for the early diagnosis of an infection. IgG is produced 10–15 d after infection and is present for a long period after the disease course has ended. Furthermore, IgG can persist in the body, even in the blood, for a lifetime. A positive IgG Ab test can indicate a pathogenic infection. When the same pathogen is encountered, the specific IgG binds to the pathogen, reducing its activity and preventing it from binding to human cells. IgG can also rapidly activate the innate immune system, shortening immune response time and avoiding or reducing the recurrence of the disease. Abs detection is a practical method for conducting an epidemiological survey after an outbreak, despite the reported 10%–15% deviation. It has high sensitivity and can serve as a valid metric for a long time after a disease is no longer present in an individual. Moreover, Abs detection can very accurately identify asymptomatic infected individuals and thereby helps determine the actual scale of the pandemic[17,18].

In a study of 173 patients, plasma samples (n = 535) were collected for the detection of total anti-SARS-CoV-2 Abs, IgG, and IgM. The dynamics of the antibody levels during disease progression were also analyzed. The seroconversion rates of IgG, IgM, and Abs were 64.7%, 82.7%, and 93.1%, respectively. The median seroconversion times of IgM, IgG, and Abs were 14 d, 12 d, and 11 d, respectively. One week after disease onset, the prevalence of antibodies in studied patients was less than 40%. From 15 d after onset, the prevalence increased starkly to 79.8% (IgG), 94.3% (IgM), and 100.0% (Abs). The detectability of RNA decreased from 66.7% (58/87) at day 7 to 45.5% (25/55) between 15 d and 39 d after the disease onset. Even in the early stages, the combined application of RNA and Abs detection can significantly improve the diagnosis rate of COVID-19 (p < 0.001)[19]. When rapid PCR testing for SARS-CoV-2 was not available, another study concluded that using rapid antigen-Abs combined testing improved detection accuracy, with a sensitivity of 91.2% and a specificity of 98.9%. The results of antigen and Abs testing can be used as a reliable substitute for PCR testing[20]. Clinical significance of the combined detection of SSA and SARS-CoV-2 RNA (Table 2).

Table 2 Clinical significance of combined detection of severe acute respiratory distress syndrome coronavirus-2-specific antibodies and RNA.
No.Antibody assay
Nucleic acid detectionClinical significance
IgM
IgG
1+++During the active infection period, the body has a certain resistance to COVID-19 (persistent IgG has been produced)
2+-+The patient may be in the middle of the SARS-CoV-2 infection. The body's immune response produces IgM antibodies early in the disease course. IgG has not yet been produced, or the IgG level has not reached the limit of detection
3-++The patient may be in the middle or late phase of the disease or may have a recurrent infection of SARS-CoV-2
4--+This is the COVID-19 "window period", which usually lasts 2 wk
5++-The patient is in the recovery phase of COVID-19. The virus has been cleared from the body, and IgM and IgG are positive. Alternatively, this may indicate that the nucleic acid test result was a false negative, and the patient is in the active phase of infection
6+--IgM positivity indicates that the patient may be in the early stage of infection. Suspicious nucleic acid test results require repeated sampling and verification
7±--This indicates that the patient is in the early stage of viral infection, and the viral load is very low. The patient is in the acute stage of COVID-19, and the body has not yet produced IgG. Alternatively, the result for IgM may have been an error caused by the presence of rheumatoid factor. One week later, the examination and diagnosis must be repeated based to evaluate for changes in IgM and IgG
8-+-The patient may have been infected with the virus in the past and has recovered; the virus has been cleared from the body. IgG can last for a long time, possibly even for life
9---The individual is healthy or in the incubation period of infection
ROLE OF ANTI-SARS-COV-2 SPECIFIC ANTIBODIES IN THE TREATMENT OF COVID-19
Therapeutic effect of convalescent plasma on COVID-19

Immunological studies have shown that Abs in CP can recognize antigenic determinants of pathogens, including those of viruses. These Abs can neutralize the virus, reduce the viral load, and prevent or cure disease. In other words, Abs can trigger and participate in a self-repairing process in the body. By providing Nabs, CP is a form of passive immunotherapy. The benefits of CP infusion in related diseases were first studied in the 20th century[21]. In the absence of specific treatments for emerging infectious diseases, CP therapy remains an important treatment method[22]. CP has been shown to be effective in treating patients with influenza A (H1N1)[23], severe acute respiratory syndrome[24], and Middle East respiratory syndrome[25].

On August 23, 2020, the United States FDA approved the emergency use of CP as a potential COVID-19 treatment. Plasma therapy, according to the FDA, can effectively reduce the severity or the duration of COVID-19 disease in some hospitalized patients, and the known potential benefits far outweigh the product's risks[26]. With the continuous emergence of SARS-CoV-2 variants, previously effective treatment methods have been called into question. Several other observational studies and randomized controlled trials have recently been reviewed by experts from the National Institutes of Health (NIH) in the United States[27-29]. In these studies, the efficacy of CP against COVID-19 was investigated in both outpatient and inpatient settings. The FDA updated the EUA of CP to treat COVID-19[30] on November 28, 2021. Only CP with high titers of SARS-CoV-2 Abs is thought to be effective in patients with COVID-19 who are immunocompromised (i.e. those who have recently received radiation, chemotherapy, or who have immunodeficiency disease) or have received immunosuppressive therapy[29], and the potential benefits to patients may outweigh the known potential risks. However, infusion of CP in immunocompetent COVID-19 patients is unlikely to be clinically beneficial, and its use in these patients is not advised.

The WHO treatment guidelines only recommend the use of CP in the research and clinical trial settings[31]. However, based on subgroup data analysis from several observational studies and randomized controlled trials[27-29], the NIH treatment guidelines still recommend that CP be used for COVID-19 patients with secondary or primary humoral immunodeficiency (e.g., with agammaglobulinemia, hematological malignancies, solid organ transplant, or other immunodeficiency)[32,33]. In a subset of immunocompromised patients, some findings suggest that CP may provide benefits such as improved survival and/or more days without organ support when compared to placebo [odds ratio 1.51; 95 percent confidence interval(CI): 0.80–2.92][29].

In clinical practice, a high titer CP with a volume of 200 mL can be considered first, and the need to increase the dose can be determined based on the individual patient scenario. Past and current studies have demonstrated real-world clinical efficacy for the treatment of COVID-19 with CP[34-36] This suggests that high-level randomized controlled trials (RCTs) should be designed in the future to investigate the potential therapeutic effects of CP in the treatment of COVID-19.

The Infectious Diseases Society of America and the FDA have also re-evaluated CP for use in outpatients at risk of progression due to the lack of antiviral drugs, as the polyclonal nature of CP can render SARS-CoV-2 variants less able to escape immune defenses[37,38].

Therapeutic effect of highly concentrated immunoglobulin on COVID-19

HIG is a high-titer Abs preparation made up of anti-SARS-CoV-2 mAbs from multiple CP donors that is more concentrated than anti-SARS-CoV-2 Specific antibodies (ASSAs) from a single donor (the SARS-CoV-2 Abs titer is several times higher)[39].The preparation may work by inhibiting the virus and altering inflammation. Varicella zoster immunoglobulin has been used for varicella post-exposure prophylaxis (PEP) in high-risk individuals, and cytomegalovirus immunoglobulin has been shown to be effective and safe for the prevention of post-transplant cytomegalovirus infection. However, clinical data on the treatment of COVID-19 with HIG are currently lacking[40].

Therapeutic effect of anti-SARS-CoV-2 specific antibodies against COVID-19

Spike protein (S), nucleocapsid protein, membrane protein, envelope protein, and auxiliary and nonstructural proteins are all encoded by the SARS-CoV-2 genome. S1 and S2 are thorn protein subtypes. S1 is binds to angiotensin-converting enzyme 2 receptors on host cells. It causes S2 to change conformation, fuses the viral and host cell membranes, and allows the virus to enter human cells[41]. Binding of spike protein by mAbs decreases SARS-CoV-2 infectivity, resulting in a therapeutic effect. Indeed, ASSAs have shown clinical efficacy in the treatment of SARS-CoV-2 infection[42].

Anti-SARS-CoV-2 monoclonal antibody for pre-exposure prophylaxis of SARS-CoV-2 infection

Experts recommend intramuscular tixagevimab plus cilgavimab (Evusheld) as SARS-CoV-2 pre-exposure prophylaxis (PrEP) for adults and adolescents (age ≥ 12 years and weight ≥ 40 kg) who have not been exposed to COVID-19. In patients with moderate to severe immunocompromise, the immune response to the COVID-19 vaccine may be insufficient, or vaccination with any available COVID-19 vaccine may not be complete[43]. Omicron mutant sensitivity to tixagevimab plus cilgavimab has been shown to be moderately reduced, but the regimen was still beneficial for PrEP prevention of infection by Omicron strains.

Anti-SARS-CoV-2 monoclonal antibodies for post-exposure prophylaxis of SARS-CoV-2 infection

Casirivimab plus imdevimab and bamlanivimab plus elesevimab are not recommended for PEP of SARS-CoV-2[42] in countries and regions where Omicron variant strains are prevalent as these strains are not susceptible to these drugs. Prior to Omicron, the FDA issued a EUA for this regimen, allowing PEP in people who are at high risk of SARS-CoV-2 infection and have a high risk of developing severe COVID-19 disease. This protocol should be made available for PEP of the individuals listed above in non-Omicron endemic countries and regions.

Anti-SARS-CoV-2 monoclonal antibodies for outpatient COVID-19 patients

To treat outpatients (age ≥ 12 years and weight ≥ 40 kg) with mild to moderate COVID-19 and high risk of clinical progression, the use of sotrovimab 500 mg as a single intravenous infusion within 10 d of symptom onset is recommended. Sotrovimab retains in vitro activity against Omicron variants and is believed to benefit Omicron-infected patients clinically[44].

Experts do not recommend casirivimab plus imdevimab or bamlanivimab plus etesevimab because Omicron is not susceptible to these drugs. However, because the FDA issued EUA prior to the Omicron epidemic, these regimens can be used in countries and regions where Omicron is not endemic. The use of cascilimab plus imdevimab has also been halted in the United States due to Omicron variant insensitivity[45]. Cascilimab 600 mg plus imdevimab 600 mg administered as a single intravenous infusion were approved by the FDA prior to the spread of Omicron and is approved for the treatment of patients with mild to moderate COVID-19. The use of this protocol is supported by results from a phase 3 double-blind, randomized, placebo-controlled trial in outpatients with mild to moderate COVID-19[46]. This regimen can still be used in places where non-Omicron variants are prevalent.

Anti-SARS-CoV-2 monoclonal antibodies for COVID-19 hospitalized patients

In one study, 583 outpatients with mild to moderate COVID-19 were studied. Two hundred ninety-one subjects were randomly assigned to receive sotrovimab (500 mg IV) and 292 subjects received placebo. On day 29, the primary endpoint was the proportion of hospital stays longer than 24 h or death from any cause. Three patients (1%) in the sotrovimab group and 21 patients (7%) in the placebo group required hospitalization or died as a result of disease progression (relative risk reduction, 85%; 97.24%CI: 44-96; P = 0.002). On day 29, 1 patient in the placebo group died after being admitted to the intensive care unit. Adverse events were reported in 17% of patients in the sotrovimab group and 19% of patients in the placebo group during the safety assessment; the incidence of serious adverse events was lower in the sotrovimab group than in the placebo group (2% and 6%, respectively). Sostovizumab reduced the risk of COVID-19 exacerbations in high-risk patients with mild to moderate COVID-19[47].

The use of bamlanivimab plus etesevimab to treat COVID-19 has been halted in the United States because the Omicron variant has poor susceptibility to this mAb in vitro[45]. Prior to the emergence of the Omicron variant, the BLAZE-1 phase 3 clinical trial demonstrated that bamlanivimab plus etesevimab benefits patients with mild to moderate COVID-19 at risk of progression to severe disease with or without hospitalization[46]. The main SARS-CoV-2 strain currently introduced to China from abroad is Omicron BA.2. Pfizer's antiviral drug Paxlovid (nimatovir plus ritonavir) is recommended for adults and adolescents aged 12–17 years old, weight ≥ 40 kg. Paxlovid is given in cases of mild disease within 5 d of onset according to China's Protocol for Prevention and Control of Novel Coronavirus Infection (10th Ed)[48].

Anti-SARS-CoV-2 monoclonal antibodies for hospitalized patients with severe or critical COVID-19

ASSA are not currently approved for use in hospitalized patients with severe COVID-19; however, these products may see use in such patients through an expanded access plan. Casirivimab plus imdevimab is conditionally recommended for COVID-19 patients with severe or critical illness, and may also benefit seronegative patients. According to the RECOVERY trial, a credible subgroup effect suggests that casirivimab plus imdevimab may reduce mortality and days requiring mechanical ventilation in patients with seronegative status[31].

Relevant literature is summarized in Table 3[49-54]. Many therapeutic mAbs that were previously effective became ineffective in the face of the Omicron variant; studies of these are not included.

Table 3 Sensitivity of Omicron variants to therapeutic monoclonal antibodies.
mAb (s)FDA EUATarget on SOmicron variant
BA.1
IC50
BA.2
IC50
Bamla/Etese Yes BRD or SReduction in activity vs control approximately 1000-fold (highly resistant)> 10000 ng/mLReduction in activity vs control approximately 1000-fold (highly resistant)> 10000 ng/mL
Casir/Imdev YesBRD Reduction in activity vs control approximately 1000-fold (highly resistant)> 10000 ng/mLReduction in activity vs control approximately 1000-fold (highly resistant)> 10000 ng/mL
SotroYes BRD Median fold reduction in susceptibility 4.0 (IQR: 2.6 to 6.9)Median 276 ng/mL (IQR: 163 to 423)Median fold reduction in susceptibility 17 (IQR: 13 to 30)Median 1250 ng/mL (IQR: 567 to 1456)
Cilag/TixagYes BRD Median fold reduction in susceptibility 86 (IQR:27 to 151). The FDA recommended that the dosage for each mAb in this combination be increased 300 mg and administered intramuscularlyMedian 256 ng/mL (IQR: 170 to 750) Median fold reduction in susceptibility 5.4 (IQR: 3.7 to 6.9). Nearly complete restoration BA.2 susceptibility to cilgavimabMedian 44 ng/mL (IQR: 27 to 73)
BebteYes BRD Median fold reduction in susceptibility 1.0 (IQR: 0.7 to 1.4) Bebtelovimab is the only mAb active against the current dominant circulating Omicron variant; in non- hospitalized adults, bebtelovimab may be used as an alternative therapy when no preferred therapy (e.g., nirmatrelvir/ritonavir, remdesivir) available Median 2.6 ng/mL (IQR: 1.8 to 5.0)Median fold reduction in susceptibility 1.0 (IQR: 0.7 to 1)Median 4.0 ng/mL (IQR: 0.8 to 5.0)
RegdaNoBRDDisplayed little residual activity NADisplayed little residual activity NA
AmubaNoBRDDisplayed little residual activity NADisplayed little residual activityNA
RomluNoBRDRetained partial activity NADisplayed little residual activityNA
AdintNoBRDRetained partial activityNANA
Safety of monoclonal antibodies in clinical application

In most studies, adverse events with the use of therapeutic mAbs were not serious (e.g., nausea, diarrhea) and were mostly self-limiting. The most common adverse events were mild reactions, chills, headache, injection site pain, and bronchospasm. Serious adverse events were rare, and observed instances of shortness of breath may be associated with exacerbations of COVID-19 itself[37].

Limitations of this article

SARS-CoV-2 has been raging for nearly 3 years. In the past 2 years, the mutation of SARS-CoV-2 has become more and more frequent, and transmission speed is also faster, especially with the Omicron variant. Fortunately, Omicron is becoming less toxic. The mutation of the virus reduces the efficacy of existing therapeutic drugs, which can now only be applied in some special circumstances. This is the case of the use of CP or mAbs in the treatment of COVID-19. The FDA's EUAs for many drugs are require constant updates. As human efforts cannot keep up with the speed of Omicron mutation, we should redouble our efforts to proactively discover the pattern of SARS-CoV-2 mutation and finally defeat the novel coronavirus.

CONCLUSION

SSA detection technology is widely used in clinical practice. It can assist PCR nucleic acid in the diagnosis, screening, and tracking of infection in patients with COVID-19 (including mild and asymptomatic infections). Thus, it is valuable for public health and has many potential clinical applications[55].

Because of the emergence of Delta and Omicron variants, the use of CP is severely limited. It is only recommended for use in COVID-19 patients with impaired immune function, immunodeficiency, or who are taking immunosuppressants. HIG could theoretically treat COVID-19, but there is currently insufficient data to guide its use. Treatment with ASSAs has made significant progress, and outpatient outcomes are promising. Several clinical trials[27-29] have assessed ASSA's efficacy and safety in the treatment of non-hospitalized COVID-19 patients. Sotrovimab was evaluated in the Phase 3 trial COMET-ICE for the treatment of COVID-19. This mAb treatment is thought to be more effective, and has proven effective in preventing COVID-19[47]. The United States FDA granted a EUA for the use of sotrovimab[56] in May 2021. The BLAZE-1 study investigated the efficacy of intravenous bamlanimab in the early treatment of COVID-19[57]. Bebtelovimab is the only anti-SARS-CoV-2 mAb active against the current dominant circulating Omicron variant[50]. Monoclonal antibodies can protect unvaccinated people or people exposed to high-risk environments in a prophylactic manner. The effectiveness of mAbs targeting the SARS-CoV-2 spike protein is also promising; however, research must continue to actively pursue new drugs and methods for the accurate and rapid diagnosis and treatment of COVID-19.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medicine, research and experimental

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): E

P-Reviewer: Kumar I, India; Nassar G, France; Wijaya JH, Indonesia S-Editor: Li L L-Editor: Filipodia P-Editor: Li L

References
1.  Aleem A, Akbar Samad AB, Slenker AK.   Emerging Variants of SARS-CoV-2 And Novel Therapeutics Against Coronavirus (COVID-19). 2022 Oct 10. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 Jan-.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Callaway E. Coronavirus variant XBB.1.5 rises in the United States - is it a global threat? Nature. 2023;613:222-223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 35]  [Reference Citation Analysis (0)]
3.  World Health Organization  Coronavirus Disease (COVID-19) Dashboard. [cited 13 January 2023]. Available from: https://covid19.who.int/.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Infantino M, Damiani A, Gobbi FL, Grossi V, Lari B, Macchia D, Casprini P, Veneziani F, Villalta D, Bizzaro N, Cappelletti P, Fabris M, Quartuccio L, Benucci M, Manfredi M. Serological Assays for SARS-CoV-2 Infectious Disease: Benefits, Limitations and Perspectives. Isr Med Assoc J. 2020;22:203-210.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Ye Q, Wang B, Mao J. The pathogenesis and treatment of the `Cytokine Storm' in COVID-19. J Infect. 2020;80:607-613.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1858]  [Cited by in F6Publishing: 1823]  [Article Influence: 455.8]  [Reference Citation Analysis (0)]
6.  National Institutes of Health  Coronavirus disease 2019 (COVID-19) treatment guidelines. 2021. [cited 15 February 2022]. Available from: www.covid19treatmentguidelines.nih.gov.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Bartoletti M, Azap O, Barac A, Bussini L, Ergonul O, Krause R, Paño-Pardo JR, Power NR, Sibani M, Szabo BG, Tsiodras S, Verweij PE, Zollner-Schwetz I, Rodríguez-Baño J. ESCMID COVID-19 living guidelines: drug treatment and clinical management. Clin Microbiol Infect. 2022;28:222-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 86]  [Article Influence: 43.0]  [Reference Citation Analysis (0)]
8.  Shankar-Hari M, Vale CL, Godolphin PJ, Fisher D, Higgins JPT, Spiga F, Savovic J, Tierney J, Baron G, Benbenishty JS, Berry LR, Broman N, Cavalcanti AB, Colman R, De Buyser SL, Derde LPG, Domingo P, Omar SF, Fernandez-Cruz A, Feuth T, Garcia F, Garcia-Vicuna R, Gonzalez-Alvaro I, Gordon AC, Haynes R, Hermine O, Horby PW, Horick NK, Kumar K, Lambrecht BN, Landray MJ, Leal L, Lederer DJ, Lorenzi E, Mariette X, Merchante N, Misnan NA, Mohan SV, Nivens MC, Oksi J, Perez-Molina JA, Pizov R, Porcher R, Postma S, Rajasuriar R, Ramanan AV, Ravaud P, Reid PD, Rutgers A, Sancho-Lopez A, Seto TB, Sivapalasingam S, Soin AS, Staplin N, Stone JH, Strohbehn GW, Sunden-Cullberg J, Torre-Cisneros J, Tsai LW, van Hoogstraten H, van Meerten T, Veiga VC, Westerweel PE, Murthy S, Diaz JV, Marshall JC, Sterne JAC; WHO Rapid Evidence Appraisal for COVID-19 Therapies (REACT) Working Group. Association Between Administration of IL-6 Antagonists and Mortality Among Patients Hospitalized for COVID-19: A Meta-analysis. JAMA. 2021;326:499-518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 434]  [Cited by in F6Publishing: 397]  [Article Influence: 132.3]  [Reference Citation Analysis (0)]
9.  Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR, Ghazaryan V, Marconi VC, Ruiz-Palacios GM, Hsieh L, Kline S, Tapson V, Iovine NM, Jain MK, Sweeney DA, El Sahly HM, Branche AR, Regalado Pineda J, Lye DC, Sandkovsky U, Luetkemeyer AF, Cohen SH, Finberg RW, Jackson PEH, Taiwo B, Paules CI, Arguinchona H, Erdmann N, Ahuja N, Frank M, Oh MD, Kim ES, Tan SY, Mularski RA, Nielsen H, Ponce PO, Taylor BS, Larson L, Rouphael NG, Saklawi Y, Cantos VD, Ko ER, Engemann JJ, Amin AN, Watanabe M, Billings J, Elie MC, Davey RT, Burgess TH, Ferreira J, Green M, Makowski M, Cardoso A, de Bono S, Bonnett T, Proschan M, Deye GA, Dempsey W, Nayak SU, Dodd LE, Beigel JH; ACTT-2 Study Group Members. Baricitinib plus Remdesivir for Hospitalized Adults with Covid-19. N Engl J Med. 2021;384:795-807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1159]  [Cited by in F6Publishing: 1178]  [Article Influence: 392.7]  [Reference Citation Analysis (1)]
10.  Marconi VC, Ramanan AV, de Bono S, Kartman CE, Krishnan V, Liao R, Piruzeli MLB, Goldman JD, Alatorre-Alexander J, de Cassia Pellegrini R, Estrada V, Som M, Cardoso A, Chakladar S, Crowe B, Reis P, Zhang X, Adams DH, Ely EW; COV-BARRIER Study Group. Efficacy and safety of baricitinib for the treatment of hospitalised adults with COVID-19 (COV-BARRIER): a randomised, double-blind, parallel-group, placebo-controlled phase 3 trial. Lancet Respir Med. 2021;9:1407-1418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 413]  [Article Influence: 137.7]  [Reference Citation Analysis (0)]
11.  Xiao AT, Gao C, Zhang S. Profile of specific antibodies to SARS-CoV-2: The first report. J Infect. 2020;81:147-178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 130]  [Article Influence: 32.5]  [Reference Citation Analysis (0)]
12.  Lee YL, Liao CH, Liu PY, Cheng CY, Chung MY, Liu CE, Chang SY, Hsueh PR. Dynamics of anti-SARS-Cov-2 IgM and IgG antibodies among COVID-19 patients. J Infect. 2020;81:e55-e58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 100]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
13.  Yong G, Yi Y, Tuantuan L, Xiaowu W, Xiuyong L, Ang L, Mingfeng H. Evaluation of the auxiliary diagnostic value of antibody assays for the detection of novel coronavirus (SARS-CoV-2). J Med Virol. 2020;92:1975-1979.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 40]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
14.  Timilsina SS, Durr N, Jolly P, Ingber DE. Rapid quantitation of SARS-CoV-2 antibodies in clinical samples with an electrochemical sensor. Biosens Bioelectron. 2023;223:115037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
15.  Hajissa K, Mussa A, Karobari MI, Abbas MA, Ibrahim IK, Assiry AA, Iqbal A, Alhumaid S, Mutair AA, Rabaan AA, Messina P, Scardina GA. The SARS-CoV-2 Antibodies, Their Diagnostic Utility, and Their Potential for Vaccine Development. Vaccines (Basel). 2022;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
16.  Zamani M, Ghasemi A, Shamshirgaran M, Ahmadpour S, Hormati A, Khodadadi J, Varnasseri M, Amini F, Shayanrad A, Younesi V, Poustchi H, Shabani M. Investigation of Durability of SARS-CoV-2-specific IgG and IgM Antibodies in Recovered COVID-19 Patients: A Prospective Study. Avicenna J Med Biotechnol. 2022;14:233-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
17.  Wu X, Fu B, Chen L, Feng Y. Serological tests facilitate identification of asymptomatic SARS-CoV-2 infection in Wuhan, China. J Med Virol. 2020;92:1795-1796.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 57]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
18.  Long QX, Liu BZ, Deng HJ, Wu GC, Deng K, Chen YK, Liao P, Qiu JF, Lin Y, Cai XF, Wang DQ, Hu Y, Ren JH, Tang N, Xu YY, Yu LH, Mo Z, Gong F, Zhang XL, Tian WG, Hu L, Zhang XX, Xiang JL, Du HX, Liu HW, Lang CH, Luo XH, Wu SB, Cui XP, Zhou Z, Zhu MM, Wang J, Xue CJ, Li XF, Wang L, Li ZJ, Wang K, Niu CC, Yang QJ, Tang XJ, Zhang Y, Liu XM, Li JJ, Zhang DC, Zhang F, Liu P, Yuan J, Li Q, Hu JL, Chen J, Huang AL. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat Med. 2020;26:845-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1982]  [Cited by in F6Publishing: 1966]  [Article Influence: 491.5]  [Reference Citation Analysis (0)]
19.  Zhao J, Yuan Q, Wang H, Liu W, Liao X, Su Y, Wang X, Yuan J, Li T, Li J, Qian S, Hong C, Wang F, Liu Y, Wang Z, He Q, Li Z, He B, Zhang T, Fu Y, Ge S, Liu L, Zhang J, Xia N, Zhang Z. Antibody Responses to SARS-CoV-2 in Patients With Novel Coronavirus Disease 2019. Clin Infect Dis. 2020;71:2027-2034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1590]  [Cited by in F6Publishing: 1765]  [Article Influence: 441.3]  [Reference Citation Analysis (0)]
20.  Mori K, Imaki S, Ohyama Y, Satoh K, Abe T, Takeuchi I. Rapid screening for severe acute respiratory syndrome coronavirus 2 infection with a combined point-of-care antigen test and an immunoglobulin G antibody test. PLoS One. 2022;17:e0263327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
21.  Marano G, Vaglio S, Pupella S, Facco G, Catalano L, Liumbruno GM, Grazzini G. Convalescent plasma: new evidence for an old therapeutic tool? Blood Transfus. 2016;14:152-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 157]  [Reference Citation Analysis (0)]
22.  Burnouf T, Seghatchian J. Ebola virus convalescent blood products: where we are now and where we may need to go. Transfus Apher Sci. 2014;51:120-125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 51]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
23.  Hung IF, To KK, Lee CK, Lee KL, Chan K, Yan WW, Liu R, Watt CL, Chan WM, Lai KY, Koo CK, Buckley T, Chow FL, Wong KK, Chan HS, Ching CK, Tang BS, Lau CC, Li IW, Liu SH, Chan KH, Lin CK, Yuen KY. Convalescent plasma treatment reduced mortality in patients with severe pandemic influenza A (H1N1) 2009 virus infection. Clin Infect Dis. 2011;52:447-456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 486]  [Cited by in F6Publishing: 473]  [Article Influence: 36.4]  [Reference Citation Analysis (0)]
24.  Soo YO, Cheng Y, Wong R, Hui DS, Lee CK, Tsang KK, Ng MH, Chan P, Cheng G, Sung JJ. Retrospective comparison of convalescent plasma with continuing high-dose methylprednisolone treatment in SARS patients. Clin Microbiol Infect. 2004;10:676-678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 271]  [Cited by in F6Publishing: 274]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
25.  Arabi YM, Hajeer AH, Luke T, Raviprakash K, Balkhy H, Johani S, Al-Dawood A, Al-Qahtani S, Al-Omari A, Al-Hameed F, Hayden FG, Fowler R, Bouchama A, Shindo N, Al-Khairy K, Carson G, Taha Y, Sadat M, Alahmadi M. Feasibility of Using Convalescent Plasma Immunotherapy for MERS-CoV Infection, Saudi Arabia. Emerg Infect Dis. 2016;22:1554-1561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 169]  [Article Influence: 28.2]  [Reference Citation Analysis (0)]
26.  United States Food and Drug Administration  Recommendations for Investigational COVID-19 Convalescent Plasma. [cited 8 February 2023]. Available from: www.fda.gov/vaccines-blood-biologics/investigational-new-drug-applications-inds-cber-regulated-products/recommendations-investigational-covid-19-convalescent-plasma.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  RECOVERY Collaborative Group. Convalescent plasma in patients admitted to hospital with COVID-19 (RECOVERY): a randomised controlled, open-label, platform trial. Lancet. 2021;397:2049-2059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 279]  [Article Influence: 93.0]  [Reference Citation Analysis (0)]
28.  Bégin P, Callum J, Jamula E, Cook R, Heddle NM, Tinmouth A, Zeller MP, Beaudoin-Bussières G, Amorim L, Bazin R, Loftsgard KC, Carl R, Chassé M, Cushing MM, Daneman N, Devine DV, Dumaresq J, Fergusson DA, Gabe C, Glesby MJ, Li N, Liu Y, McGeer A, Robitaille N, Sachais BS, Scales DC, Schwartz L, Shehata N, Turgeon AF, Wood H, Zarychanski R, Finzi A; CONCOR-1 Study Group, Arnold DM. Convalescent plasma for hospitalized patients with COVID-19: an open-label, randomized controlled trial. Nat Med. 2021;27:2012-2024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 171]  [Article Influence: 57.0]  [Reference Citation Analysis (0)]
29.  Estcourt LJ, Turgeon AF, McQuilten ZK, McVerry BJ, Al-Beidh F, Annane D, Arabi YM, Arnold DM, Beane A, Bégin P, van Bentum-Puijk W, Berry LR, Bhimani Z, Birchall JE, Bonten MJM, Bradbury CA, Brunkhorst FM, Buxton M, Callum JL, Chassé M, Cheng AC, Cove ME, Daly J, Derde L, Detry MA, De Jong M, Evans A, Fergusson DA, Fish M, Fitzgerald M, Foley C, Goossens H, Gordon AC, Gosbell IB, Green C, Haniffa R, Harvala H, Higgins AM, Hills TE, Hoad VC, Horvat C, Huang DT, Hudson CL, Ichihara N, Laing E, Lamikanra AA, Lamontagne F, Lawler PR, Linstrum K, Litton E, Lorenzi E, MacLennan S, Marshall J, McAuley DF, McDyer JF, McGlothlin A, McGuinness S, Miflin G, Montgomery S, Mouncey PR, Murthy S, Nichol A, Parke R, Parker JC, Priddee N, Purcell DFJ, Reyes LF, Richardson P, Robitaille N, Rowan KM, Rynne J, Saito H, Santos M, Saunders CT, Serpa Neto A, Seymour CW, Silversides JA, Tinmouth AA, Triulzi DJ, Turner AM, van de Veerdonk F, Walsh TS, Wood EM, Berry S, Lewis RJ, Menon DK, McArthur C, Zarychanski R, Angus DC, Webb SA, Roberts DJ, Shankar-Hari M; Writing Committee for the REMAP-CAP Investigators. Effect of Convalescent Plasma on Organ Support-Free Days in Critically Ill Patients With COVID-19: A Randomized Clinical Trial. JAMA. 2021;326:1690-1702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 132]  [Article Influence: 44.0]  [Reference Citation Analysis (0)]
30.  Food and Drug Administration  EUA of COVID-19 convalescent plasma for the treatment of COVID-19 in hospitalized patients: fact sheet for health care providers. 2021. [sited 15 February 2023]. Available from: https://www.fda.gov/media/141478/download.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Lamontagne F, Agarwal A, Rochwerg B, Siemieniuk RA, Agoritsas T, Askie L, Lytvyn L, Leo YS, Macdonald H, Zeng L, Amin W, da Silva ARA, Aryal D, Barragan FAJ, Bausch FJ, Burhan E, Calfee CS, Cecconi M, Chacko B, Chanda D, Dat VQ, De Sutter A, Du B, Freedman S, Geduld H, Gee P, Gotte M, Harley N, Hashimi M, Hunt B, Jehan F, Kabra SK, Kanda S, Kim YJ, Kissoon N, Krishna S, Kuppalli K, Kwizera A, Lado Castro-Rial M, Lisboa T, Lodha R, Mahaka I, Manai H, Mendelson M, Migliori GB, Mino G, Nsutebu E, Preller J, Pshenichnaya N, Qadir N, Relan P, Sabzwari S, Sarin R, Shankar-Hari M, Sharland M, Shen Y, Ranganathan SS, Souza JP, Stegemann M, Swanstrom R, Ugarte S, Uyeki T, Venkatapuram S, Vuyiseka D, Wijewickrama A, Tran L, Zeraatkar D, Bartoszko JJ, Ge L, Brignardello-Petersen R, Owen A, Guyatt G, Diaz J, Kawano-Dourado L, Jacobs M, Vandvik PO. A living WHO guideline on drugs for covid-19. BMJ. 2020;370:m3379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 475]  [Cited by in F6Publishing: 475]  [Article Influence: 118.8]  [Reference Citation Analysis (0)]
32.  Ferrari S, Caprioli C, Weber A, Rambaldi A, Lussana F. Convalescent hyperimmune plasma for chemo-immunotherapy induced immunodeficiency in COVID-19 patients with hematological malignancies. Leuk Lymphoma. 2021;62:1490-1496.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 21]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
33.  Senefeld JW, Klassen SA, Ford SK, Senese KA, Wiggins CC, Bostrom BC, Thompson MA, Baker SE, Nicholson WT, Johnson PW, Carter RE, Henderson JP, Hartman WR, Pirofski LA, Wright RS, Fairweather L, Bruno KA, Paneth NS, Casadevall A, Joyner MJ. Use of convalescent plasma in COVID-19 patients with immunosuppression. Transfusion. 2021;61:2503-2511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 42]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
34.  Kanj S, Al-Omari B. Convalescent Plasma Transfusion for the Treatment of COVID-19 in Adults: A Global Perspective. Viruses. 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
35.  O'Donnell MR, Grinsztejn B, Cummings MJ, Justman JE, Lamb MR, Eckhardt CM, Philip NM, Cheung YK, Gupta V, João E, Pilotto JH, Diniz MP, Cardoso SW, Abrams D, Rajagopalan KN, Borden SE, Wolf A, Sidi LC, Vizzoni A, Veloso VG, Bitan ZC, Scotto DE, Meyer BJ, Jacobson SD, Kantor A, Mishra N, Chauhan LV, Stone EF, Dei Zotti F, La Carpia F, Hudson KE, Ferrara SA, Schwartz J, Stotler BA, Lin WW, Wontakal SN, Shaz B, Briese T, Hod EA, Spitalnik SL, Eisenberger A, Lipkin WI. A randomized double-blind controlled trial of convalescent plasma in adults with severe COVID-19. J Clin Invest. 2021;131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 112]  [Article Influence: 37.3]  [Reference Citation Analysis (0)]
36.  Kloypan C, Saesong M, Sangsuemoon J, Chantharit P, Mongkhon P. CONVALESCENT plasma for COVID-19: A meta-analysis of clinical trials and real-world evidence. Eur J Clin Invest. 2021;51:e13663.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 20]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
37.  Focosi D, McConnell S, Casadevall A, Cappello E, Valdiserra G, Tuccori M. Monoclonal antibody therapies against SARS-CoV-2. Lancet Infect Dis. 2022;22:e311-e326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 91]  [Article Influence: 45.5]  [Reference Citation Analysis (0)]
38.  Infectious Diseases Society of America  Infectious Diseases Society of America IDSA guidelines on the treatment and management of patients with COVID-19.2022. [sited 8 January 2023]. Available from: https://www.idsociety.org/practice-guideline/covid-19-guideline-treatment-and-management/.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Vandeberg P, Cruz M, Diez JM, Merritt WK, Santos B, Trukawinski S, Wellhouse A, Jose M, Willis T. Production of anti-SARS-CoV-2 hyperimmune globulin from convalescent plasma. Transfusion. 2021;61:1705-1709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
40.  National Institutes of Health  Immunoglobulins: SARS-CoV-2 Specific. [sited 26 September 2022]. Available from: https://www.covid19treatmentguidelines.nih.gov/search/?q=Immunoglobulins%3A%20SARS-CoV-2%20Specific.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Jiang S, Hillyer C, Du L. Neutralizing Antibodies against SARS-CoV-2 and Other Human Coronaviruses. Trends Immunol. 2020;41:355-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 483]  [Cited by in F6Publishing: 573]  [Article Influence: 143.3]  [Reference Citation Analysis (0)]
42.  National Institutes of Health  Anti-SARS-CoV-2 Monoclonal Antibodies. [sited 24 September 2022] Available from: https://www.covid19treatmentguidelines.nih.gov/search/?q=Anti-SARS-CoV-2%20Monoclonal%20Antibodies.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Food and Drug Administration  Fact sheet for healthcare providers: emergency use authorization for Evusheld (tixagevimab co-packaged with cilgavimab). 2021. [sited 8 January 2023]. Available from: https://www.fda.gov/media/154701/download.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Food and Drug Administration  Fact sheet for healthcare providers: emergency use authorization (EUA) of sotrovimab. 2021. [sited 8 January 2023]. Available from: https://www.fda.gov/media/149534/download.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Food and Drug Administration  Coronavirus (COVID-19) | Drugs. Dec 22, 2022. [sited 8 January 2023]. Available from: https://www.fda.gov/drugs/emergency-preparedness-drugs/coronavirus-covid-19-drugs#:~:text=Sars-Cov-2-Targeting%20Monoclonal%20Antibodies.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Dougan M, Azizad M, Mocherla B, Gottlieb RL, Chen P, Hebert C, Perry R, Boscia J, Heller B, Morris J, Crystal C, Igbinadolor A, Huhn G, Cardona J, Shawa I, Kumar P, Blomkalns A, Adams AC, Van Naarden J, Custer KL, Knorr J, Oakley G, Schade AE, Holzer TR, Ebert PJ, Higgs RE, Sabo J, Patel DR, Dabora MC, Williams M, Klekotka P, Shen L, Skovronsky DM, Nirula A. A Randomized, Placebo-Controlled Clinical Trial of Bamlanivimab and Etesevimab Together in High-Risk Ambulatory Patients With COVID-19 and Validation of the Prognostic Value of Persistently High Viral Load. Clin Infect Dis. 2022;75:e440-e449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 35]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
47.  Gupta A, Gonzalez-Rojas Y, Juarez E, Crespo Casal M, Moya J, Falci DR, Sarkis E, Solis J, Zheng H, Scott N, Cathcart AL, Hebner CM, Sager J, Mogalian E, Tipple C, Peppercorn A, Alexander E, Pang PS, Free A, Brinson C, Aldinger M, Shapiro AE; COMET-ICE Investigators. Early Treatment for Covid-19 with SARS-CoV-2 Neutralizing Antibody Sotrovimab. N Engl J Med. 2021;385:1941-1950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 592]  [Cited by in F6Publishing: 664]  [Article Influence: 221.3]  [Reference Citation Analysis (0)]
48.  National Health Commission of China  Novel Coronavirus Pneumonia Diagnosis and Treatment Plan (Trial Version 9). Mar 3, 2022. [cited 2 April 2022]. Available from: http://www.gov.cn/zhengce/zhengceku/2022-03/15/5679257/files/49854a49c7004f4ea9e622f3f2c568d8.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Tao K, Tzou PL, Kosakovsky Pond SL, Ioannidis JPA, Shafer RW. Susceptibility of SARS-CoV-2 Omicron Variants to Therapeutic Monoclonal Antibodies: Systematic Review and Meta-analysis. Microbiol Spectr. 2022;10:e0092622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 35]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
50.  National Institutes of Health  Coronavirus Disease 2019 (COVID-19) Treatment Guidelines. [sited: 28 September 2022]. Available from: https://www.covid19treatmentguidelines.nih.gov/.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  United States Food and Drug Administration  Fact sheet for healthcare providers: emergency use authorization for bebtelovimab. 2022. [sited 8 January 2023]. Available from: https://www.fda.gov/media/156152/download.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  United States Food and Drug Administration  Fact sheet for healthcare providers: emergency use authorization (EUA) of sotrovimab. 2022. [sited 8 January 2023] Available from: https://www.fda.gov/media/149534/download.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  United States Food and Drug Administration  Fact sheet for healthcare providers: emergency use authorization for Evusheld (tixagevimab co-packaged with cilgavimab). 2022. [sited 8 January 2023]. Available from: https://www.fda.gov/media/154701/download.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Dong J, Zost SJ, Greaney AJ, Starr TN, Dingens AS, Chen EC, Chen RE, Case JB, Sutton RE, Gilchuk P, Rodriguez J, Armstrong E, Gainza C, Nargi RS, Binshtein E, Xie X, Zhang X, Shi PY, Logue J, Weston S, McGrath ME, Frieman MB, Brady T, Tuffy KM, Bright H, Loo YM, McTamney PM, Esser MT, Carnahan RH, Diamond MS, Bloom JD, Crowe JE Jr. Genetic and structural basis for SARS-CoV-2 variant neutralization by a two-antibody cocktail. Nat Microbiol. 2021;6:1233-1244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 166]  [Article Influence: 55.3]  [Reference Citation Analysis (0)]
55.  Zhen-Dong Y, Gao-Jun Z, Run-Ming J, Zhi-Sheng L, Zong-Qi D, Xiong X, Guo-Wei S. Clinical and transmission dynamics characteristics of 406 children with coronavirus disease 2019 in China: A review. J Infect. 2020;81:e11-e15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 43]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
56.  United States Food and Drug Administration  Coronavirus (COVID-19) Update: FDA Authorizes New Monoclonal Antibody for Treatment of COVID-19 that Retains Activity Against Omicron Variant. Feb 11, 2022. [stied 12 February 2023]. Accessed from: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-new-monoclonal-antibody-treatment-covid-19-retains.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Chen P, Nirula A, Heller B, Gottlieb RL, Boscia J, Morris J, Huhn G, Cardona J, Mocherla B, Stosor V, Shawa I, Adams AC, Van Naarden J, Custer KL, Shen L, Durante M, Oakley G, Schade AE, Sabo J, Patel DR, Klekotka P, Skovronsky DM; BLAZE-1 Investigators. SARS-CoV-2 Neutralizing Antibody LY-CoV555 in Outpatients with Covid-19. N Engl J Med. 2021;384:229-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1009]  [Cited by in F6Publishing: 944]  [Article Influence: 314.7]  [Reference Citation Analysis (0)]