eISSN: 2300-6722
ISSN: 1899-1874
Medical Studies/Studia Medyczne
Bieżący numer Archiwum Artykuły zaakceptowane O czasopiśmie Suplementy Rada naukowa Bazy indeksacyjne Prenumerata Kontakt Zasady publikacji prac
Panel Redakcyjny
Zgłaszanie i recenzowanie prac online
3/2023
vol. 39
 
Poleć ten artykuł:
Udostępnij:
Artykuł przeglądowy

COVID-19 – odpowiedź humoralna

Adam Tworek
1
,
Andrzej Rydzewski
2, 3
,
Grażyna Rydzewska
1, 4
,
Martyna Głuszek-Osuch
1, 4
,
Konrad Lewandowski
1

1.
Clinical Department of Internal Medicine and Gastroenterology with Inflammatory Bowel Disease Unit, Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
2.
Department of Internal Medicine, Nephrology and Transplantation Medicine, Central Clinical Hospital of the Ministry of Interior and Administration, Warsaw, Poland
3.
Centre of Postgraduate Medical Education, Warsaw, Poland
4.
Collegium Medicum, Jan Kochanowski University, Kielce, Poland
Medical Studies/Studia Medyczne 2023; 39 (3): 296–303
Data publikacji online: 2023/09/30
Plik artykułu:
- COVID-19 humoral.pdf  [0.32 MB]
Pobierz cytowanie
 
Metryki PlumX:
 

Introduction

The SARS-CoV-2 virus, which caused the COVID-19 pandemic, has been the subject of a vast number of studies around the world. In this article, we review current studies regarding SARS-CoV-2 antibodies, including neutralizing antibodies (NAbs) and post-vaccination response. According to Pekar et al. [1], the first human-to-human transmission took place in mid-October or mid-November 2019. The World Health Organization (WHO) announced a public health emergency of international concern on January 30, 2020 and the COVID-19 pandemic was declared on March 11, 2020. By July 2020, more than 300,000 people had died because of COVID-19 and the virus had spread around the world. We witnessed a very rapid reaction by the scientific community: by January 5, 2020, the Wuhan Institute of Virology had sequenced the whole virus genome [2], starting the large-scale race for a COVID-19 vaccine. This sequencing took place just one week after the first case, which was reported on December 31, 2019. The first COVID-19 human vaccine trials began in March 2020. After numerous studies, the BNT162b2 vaccine was finally allowed for commercial use on December 2, 2020 – initially by the United Kingdom’s Medicines and Healthcare products Regulatory Agency (MHRA), and subsequently by other countries’ regulatory agencies. More than one billion COVID-19 vaccine doses had been administered globally by April 2021.
The SARS-CoV-2 virus is closely related to two highly pathogenic coronaviruses: severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). The novel coronavirus shares 79% of its genome sequence with SARS-CoV and 50% with MERS-CoV [3]. Despite their differences, these viruses have one thing in common: they have jumped the immunology barrier and started human-to-human transmission.
Antibodies (along with dendritic cells, T cells, and B cells) play a crucial role in communicating the presence of a pathogen to immune effector cells. An antibody, also known as immunoglobin (Ig), is a heavy, Y-shaped protein used to identify and destroy pathogens such as bacteria and viruses. Most antibodies are composed of four polypeptide chains – two copies of a heavy chain and two copies of a light chain – connected by disulfide bonds [4]. Moreover, an antibody is also separated into two antigen-binding fragments (FAB) contacting domains and forming a characteristic Y shape. The FV region is the subregion of FAB that binds to an antigen. There are two identical antibody-binding sites on a single antigen, which allows it to bind strongly to a multivalent antigen and to form antibody complexes. This plays a crucial role in activating other parts of the immune system. Being aware of this virus’s specific antibodies could facilitate efforts to end this and future pandemics quickly.

SARS-CoV-2 structure and mechanism of entry into cells

The SARS-CoV-2 virus is built of various proteins: membrane (M), envelope (E), nucleocapsid (N), and spike (S). The N protein holds the virus’s RNA genome. Together, the E, S, and M proteins create the viral envelope. The mechanism of entry into human cells is closely related to the angiotensin-converting enzyme 2 receptor (ACE2) [5, 6]. Coronaviruses use the homotrimeric spike glycoprotein, which is divided into S1 and S2 subunits on the envelope, to bind to their receptors. SARS-CoV-2 enters human cells with the mediation of the spike protein, which is duplicated multiple times on the surface of the virus. The S1 subunit binds to the ACE2 receptor, while the S2 subunit binds to the cell membrane, initiating peptide-mediating membrane fusion. The mechanism of SARS-CoV-2 membrane fusion is very similar to that proposed for HIV membrane fusion [7]. At this stage, the first cellular immune mechanisms can be observed. Toll-like receptors (TLRs) are responsible for producing type I interferons once they recognize the virus [8]. Among these TLRs, TLR7 and TLR8 play a special role: they bind single-stranded viral RNA and induce pro- inflammatory cytokines. These two TLRs are expressed mostly in the lung, brain, skin, and lymphoid tissues.

Antibody response in COVID-19 patients

In the early stage of the pandemic, Zhang et al. were the first to establish a specific SARS-CoV-2 antibody detection test [9]. Moreover, they noted that a molecular diagnosis based on oral swabs detected only 50% of COVID-19 cases; in contrast, the serology test detected almost 100% of the positive cases. After infection, the S and N proteins are the main targets of the immune response. According to Sun et al., the antibody response was more chaotic and less linear in ICU patients than in non-ICU patients [10]. N protein antibodies, unlike the S protein antibodies, are unable to neutralize the virus [11]. NAbs interact with other parts of the immune system, such as phagocytes or natural killer cells, facilitating effective neutralization of SARS-CoV-2. Apart from their protective function, these antibodies can induce a harmful antibody-dependent enhancement (ADE), which was observed in the previous SARS-COV pandemic. ADE is mediated by the Fc receptors (FcRs) found on many immune cells, such as monocytes, macrophages, and B cells [12]. Furthermore, during the SARS-CoV pandemic, ADE in macrophages led to elevated levels of tumor necrosis factor and IL-6 [13]. Another conclusion drawn from the SARS-CoV pandemic is the fact that antibodies targeting different S-protein epitopes have different potential to induce ADE. Antibodies targeted to the receptor-binding domain (RBD) or the H2 domain of the S protein induced a better antibody response in non-human primates, while antibodies targeting other S protein domains were related to a higher risk of ADE. Moreover, recent studies have found a correlation between antibody kinetics and disease severity. Rijkers et al. observed that patients with a severe form of the disease developed a robust antibody response to SARS-CoV-2, including IgG and IgA antibodies. Patients with mild symptoms experienced a modest antibody response [14]. Ren et al. also reported a clear link between the dynamics of antibody production and disease severity. A delayed antibody response was associated with a more severe course of the disease, in contrast to the earlier response characteristic of patients with a mild form of the disease [15].

Seroconversion and antibody kinetics

A longitudinal prospective study of 1000 patients conducted by Yadav et al. reported that the median time for seroconversion was 10 days for IgG and 8 days for IgM [16]. Severe cases of COVID-19 tended to have an earlier seroconversion than mild ones. According to other studies, the combination of SARS-CoV-2 IgG and IgM antibodies was observed in 75% of patients; IgM peaked in the second week of infection, while IgG peaked in the third week [17]. This seroconversion rate differs from other studies, which reported values between 70% and 90%. This is most likely related to different laboratory methods. Zhang et al. found that among ICU patients, the seroconversion time for both IgG and IgM antibodies was up to one month [18]. According to Agarwal et al., there was no difference in seroconversion rate according to age, comorbidities, or treatment groups [19]. No correlation between antibody levels and sex was proven. The duration that antibodies are present among convalescents is a very important issue from a public health perspective. According to Hedges et al., 50% of subjects maintained a high level of neutralizing antibodies for at least 6 months. The participants with the highest starting titers had the longest-lasting response, up to 12 months after the diagnosis. In this group, the antibody response after SARS-CoV-2 vaccination was much higher and lasted longer than in the convalescent patients [20]. Achiron et al. reported that a protective level of NAbs lasted at least 9 months in a group of convalescents [21]. According to most studies, anti-spike protein and anti-RBD antibodies showed the highest neutralizing potential [22]. The available data indicate that NAbs are produced by many B cells with little or no affinity maturation required [23–25]. These data also suggest that SARS-CoV-2 NAbs are developed from naïve B cells instead of pre-existing cross-reactive memory B cells. Although there is no evidence of a correlation between age and NAb levels, Varona et al. observed that among patients older than 45 years, NAb levels were more stable than among younger patients [26].

COVID-19 antibody tests

The rapid spread of SARS-CoV-2 in society created an urgent need to develop fast diagnostic methods. Antibody tests seem to be a great alternative to expensive, time-consuming PCR methods. Even though antibody tests do not detect active disease, they do have some advantages over nucleic acid tests and antigen tests. First of all, they have a much longer detection window. Another advantage is the fact that the distribution of antibodies in the blood is more uniform than the viral load in respiratory samples, which may cause false negatives with PCR. Antibody tests may be applied in convalescent plasma donor research to determine the level of protection in patients with immune deficits, or it can simply be supplementary in the diagnosis of COVID-19. Currently, neutralization assays seem to be the standard method for these measurements. Moreover, the role of this test is to evaluate the diagnostic performance of binding antibody tests, which is the most popular antibody test method. Binding antibody tests have been developed to detect immunoglobins IgA, IgM, and IgG, including ELISA, ICG assay, electrochemiluminescence immunoassay (ECLIA), CLIA with enzyme or non-enzyme labels, fluorescence immunoassay (FIA), LFIA, protein microarrays, biosensors, and immunofluorescence assays (IFA) [27]. There are a few reasons for false positives, one of which is the cross-reactivity towards the SARS-CoV, MERS-CoV, HCoV-HKU1, HCoV-OC43, HCoV-229E, HCoV-Alpha1, dengue, and hepatitis B viruses [28–34] (Tables 1, 2).

Immune response after COVID-19 vaccination

According to the literature, the BNT162b2 vaccine induces a moderate level of anti-S1 IgA and IgM after two doses, though a strong IgG response was observed [35]. Wheeler et al. investigated the dynamics of the antibody response by comparing four vaccinations. The first dose of vaccination induced an immune response two weeks after injection. A second dose resulted in a 100-fold increase in antibodies in every group, including the patients who had not responded after the first dose. Their antibody levels were significantly higher than those of the convalescent group. According to Martinez-Baz et al., the incidence of symptomatic COVID-19 infection was three times lower in the patients vaccinated with the second dose vs. patients vaccinated only once (15.5% vs. 5.1%) [36]. Interestingly, RBD and S1 antibody levels were much higher after the second dose than any levels after the first dose. This could mean that the first dose of vaccination could induce an immunologic response, even among patients who exhibited no increase in antibodies after the first vaccination. According to Ibarrondo et al., a sharp increase in antibodies was observed in convalescents after the first dose, but there was no further rise after the second one [37]. Tada et al. observed lower seroconversion related to SARS-CoV-2 vaccination during anti-CD20 monoclonal antibody and fingolimod therapy in a group of patients with multiple sclerosis [38]. A weaker post-vaccination immunologic response was also observed in a group of patients with active cancer [39]. Moreover, it was also observed that the dynamics of falling antibody levels was similar in convalescents and vaccine recipients, with a 90% loss within 90 days [40].
Tada et al. compared NAb titers after vaccination with the mRNA vaccines BNT162b2 and mRNA-1273 versus adenoviral vector-based single-dose Ad26.COV2.S vaccination. The study reported much higher neutralization titers in the mRNA-based vaccines [38]. However, some recent studies have demonstrated that combining ChAdOx1nCoV-19 adenoviral vector vaccine with the previous BNT162b2 proved to be effective against the new variants of concern [40–43].
An important question is how the new variants of the virus affect the ability to produce specific NAbs. Muik et al. found that after two doses of BNT162b2 vaccine, Omicron-neutralizing titers were reduced 22-fold compared to the primary version of SARS-CoV-2 [41]. The level of NAbs significantly increased one month after a third dose of vaccination. This could mean that there is a need for a third dose, as well as for vaccines adapted to newer variants of this coronavirus. Khoury et al. reported that the neutralization level required for 50% protection against detectable SARS-CoV-2 was 20.2% of the mean convalescent level in seven different cohorts [42]. Larger decreases in NAb titers were reported among younger patients, those previously vaccinated against influenza and those with no previous COVID-19 infection [44–51] (Table 3).

Neutralizing antibody therapies for COVID-19

The COVID-19 pandemic has compelled society to explore new technologies to treat active infections. Drugs based on neutralizing antibodies appear to be a promising avenue. The first antibody-based drug granted emergency use authorization (EUA) in the USA was tixagevimab/cilgavimab, a combination of monoclonal antibodies administered via intramuscular injection for pre-exposure prophylaxis. In the group of patients receiving this treatment, a lower mortality rate was observed (9% compared to 12% in the placebo group). However, in February 2023, the FDA revoked the authorization due to its inefficacy against the XBB.1.5 subvariant [52]. A phase 3 study of casirivimab/imdevimab reported a 70% reduction in hospitalizations when administered at a daily dose of 1200 mg and a median duration of symptoms that was four days shorter. Despite these findings, the WHO advised against the use of casirivimab/imdevimab in the 13th version of the COVID-19 Therapeutic Guidelines, released on January 13, 2023 [53].
The most significant challenge for neutralizing antibody therapies in treating COVID-19 patients is the emergence of viral variants. The increased resistance of new SARS-CoV-2 variants, such as Omicron, to neutralizing antibodies limits the long-term development of this treatment approach. A potential solution could involve targeting conserved sites less likely to mutate. Zhou et al. reported that targeting the S2 stem-helix fusion region of the spike protein yielded comparable effectiveness while reducing the risk of mutation at this epitope [54]. Our experience treating COVID-19 with neutralizing antibodies provides valuable insight that may be highly applicable in addressing the inevitable future pandemics. The lessons learned from developing and implementing antibody-based therapies can inform and expedite the creation of effective treatments for emerging infectious diseases.

Pathological antibody response

Although the humoral response is aimed at combating the SARS-CoV-2 virus, unwanted reactions have also been observed. It has long been known that NAbs activate specific immune complexes, leading to cytokine storm [55]. Recent studies have demonstrated that an IgG-specific immune complex was detected in the vascular walls of the lungs in 6 severe cases [56]. These specific IgG complexes are associated with acute respiratory distress syndrome in the most severe cases. Some publications have reported incorrect glycosylation in the Fc region of S1/S2 IgG antibodies in critical patients [57, 58]. This incorrect glycosylation in immune complexes can lead to the excitation of macrophages with FcRIIIa or FcRIIa, which in turn can lead to cytokine storm and the secretion of cytokines such as IL-6 or tumor necrosis factor. Another unwanted effect caused by IgG-specific immune complexes is platelet-mediated thrombosis [59]. According to Bastard et al., at the onset of critical disease, 101 of 987 patients with life-threatening COVID-19 pneumonia had neutralizing IgG autoantibodies against interferon-w (13 patients), against the 13 types of IFN-a (36 patients), or against both (52 patients) [59]. For comparison, those autoantibodies were detected in only 4 out of 1227 healthy individuals. Other studies have also reported that autoantibodies against immunomodulatory proteins such as cytokines, chemokines, and cell-surface proteins have been observed in severe COVID-19 patients. This may indicate a significant role for the proteins in critical COVID-19 pathogenesis. Membrane attack complexes have been found in the liver, kidney, hearts, and lungs of patients with severe disease. SARS-CoV-2 can induce the complement activation pathway in a few ways: classical, lectin, and alternative pathways. Higher levels of IgG1 and IgG3 were found, as were higher levels of the activated complement components C3a, C3b, C4b, and C5a. This concentration of the complement activation pathway was related to disease severity [60].

Conclusions

The humoral response against SARS-CoV-2 presented in this article is just a part of the immune response, which includes cell-mediated immunity and the innate immune system. Our knowledge about this pathogen is growing every day. On the other hand, its ability to mutate rapidly is a serious global public health threat. We still need additional studies, as this knowledge may be crucial in future global pandemics as well.

Conflict of interest

The authors declare no conflict of interest.
References
1. Pekar J, Zhang Y, Gearing M, Pan M, Yan H, Zhang S, Wang G, Chen X, Chen L, Chen Z, Zhu H. Timing the SARS-CoV-2 index case in Hubei Province. Science 2021; 372: 412-417.
2. Brant AC, Simões E Silva AC, Souza JP, Abreu BJ, Duar- te AJ, Oliveira SC. SARS-CoV-2: from its discovery to genome structure, transcription, and replication. Cell Biosci 2021; 11: 211.
3. Lu R, Zhao X, Li J, Niu P, Yang B, Wu H, Wang W, Song H, Huang B, Zhu N, Bi Y. Genomic characterization and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet 2020; 395: 565-574.
4. Woof JM, Burton DR. Human antibody-Fc receptor interactions illuminated by crystal structures. Nat Rev Immunol 2004; 4: 89-99.
5. Shang J, Ye G, Shi K, Wan Y, Luo C, Aihara H, Geng Q, Auerbach A, Li F. Structural basis of receptor recognition by SARS-CoV-2. Nature 2020; 581: 221-224.
6. Lan J, Ge J, Yu J, Shan S, Zhou H, Fan S, Zhang Q, Shi X, Wang Q, Zhang L, Wang X. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 2020; 581: 215-220.
7. Shaik MM, Peng H, Lu J, Rits-Volloch S, Xu C, Liao M, Chen B. Structural basis of coreceptor recognition by HIV-1 envelope spike. Nature 2019; 565: 318-323.
8. Jangra S, De Vrieze J, Choi A, Rathnasinghe R, Laghlali G, Uvyn A, Van Herck S, Ozorowski G, De Waele J, Sche- pens W, Roose K. Sterilizing immunity against SARS-CoV-2 infection in mice by a single-shot and lipid amphiphile imidazoquinoline TLR7/8 agonist-adjuvanted recombinant spike protein vaccine. Angew Chem Int 2021; 60: 9467-9473.
9. Zhang W, Du RH, Li B, Zheng XS, Yang XL, Hu B, Wang YY, Xiao GF, Yan B, Shi ZL, Zhou P. Molecular and serological investigation of 2019-nCoV infected patients: implication of multiple shedding routes. J Med Virol 2020; 92: 1691-1697.
10. Sun B, Feng Y, Mo X, Zheng P, Wang Q, Li P, Peng P, Liu X, Chen Z, Huang H, Zhang F, Luo W, Niu X, Hu P, Wang L, Peng H, Huang Z, Feng L, Li F, Zhang F, Li F, Zhong N, Chen L. Kinetics of SARS-CoV-2 specific IgM and IgG responses in COVID-19 patients. Emerg Microbes Infect 2020; 9: 940-948.
11. Shrock E, Fujimura E, Kula T, Timms RT, Lee IH, Leng Y, Robinson ML, Sie BM, Li MZ, Chen Y, Logue J, Zuiani A, McCulloch D, Lelis FJN, Henson S, Monaco D, Tra- vers M, Habibi S, Clarke WA, Caturegli P, Laeyendecker O, Piechocka-Trocha A, Li JZ, Khatri A, Chu HY; MGH COVID-19 Collection & Processing Team; Villani AC, Kays K, Goldberg MB, Hacohen N, Filbin MR, Yu XG, Walker BD, Wesemann DR, Larman HB, Lederer JA, Elledge SJ. Viral epitope profiling of COVID-19 patients reveals cross-reactivity and correlates of severity. Science 2020; 370: 4250.
12. Iwasaki A, Yang Y. The potential danger of suboptimal antibody responses in COVID-19. Nat Rev Immunol 2020; 20: 339-341.
13. Wang SF, Tseng SP, Yen CH, Yang JY, Tsao CH, Shen CW, Chen KH, Liu FT, Liu WT, Chen YMA, Huang JC. Antibody-dependent SARS coronavirus infection is mediated by antibodies against spike proteins. Biochem Biophys Res Commun 2014; 451: 208-214.
14. Rijkers G, Murk JL, Wintermans B, van Looy B, van den Berge M, Veenemans J, Stohr J, Reusken C, van der Pol P, Reimerink J. Differences in antibody kinetics and functionality between severe and mild SARS-CoV-2 infections. J Infect Dis 2020; 222: 1265-1269.
15. Ren L, Zhang R, Chang D, Wang J, Hu Y, Chen H, Guo L, Wu C, Wang C, Wang Y, Wang Y, Wang G, Yang S, Dela Cruz CS, Sharma L, Wang L, Zhang D, Wang J. The kinetics of humoral response and its relationship with the disease severity in COVID-19. Commun Biol 2020; 3: 780.
16. Yadav AK, Ghosh S, Kotwal A, Kaushik SK, Bobdey S, Sahu R, Kapoor S, Faujdar DS, Teli PT, Anand V. Seroconversion among COVID-19 patients admitted in a dedicated COVID hospital: a longitudinal prospective study of 1000 patients. Med J Armed Forces India 2021; 77 (Suppl 1): S379-S384.
17. Sun B, Feng Y, Mo X, Zheng P, Wang Q, Li P, Peng P, Liu X, Chen Z, Huang H, Zhang F, Luo W, Niu X, Hu P, Wang L, Peng H, Huang Z, Feng L, Li F, Zhang F, Li F, Zhong N, Chen L. Kinetics of SARS-CoV-2 specific IgM and IgG responses in COVID-19 patients. Emerg Microbes Infect 2020; 9: 940-948.
18. Zhang B, Yue D, Wang Y, Wang F, Wu S, Hou H. The dynamics of immune response in COVID-19 patients with different illness severity. J Med Virol 2021; 93: 1070-1077.
19. Agarwal A, Baghmar S, Qureshi S, Khurana A, Setia R, Khan MA, Barman P, Kapoor RK, Hasthavaram NR, Kumar R, Singh N, Sarin A, Handoo A. Persistent antibody responses to SARS-CoV-2 infection in cancer patients: a single-center retrospective observational study. Indian J Med Paediatr Oncol 2021; 42: 123-129.
20. Hedges JF, Thompson MA, Snyder DT, Robison A, Tay- lor MP, Jutila MA. Titers, prevalence, and duration of Sars-Cov-2 antibodies in a local Covid-19 Outbreak and following vaccination. Vaccines 2021; 9: 587.
21. Achiron A, Gurevich M, Falb R, Dreyer-Alster S, Sonis P, Mandel M. SARS-CoV-2 antibody dynamics and B-cell memory response over time in COVID-19 convalescent subjects. Clin Microbiol Infect 2021; 27: 1349.e1-1349.e6.
22. Garcia-Beltran WF, Lam EC, Astudillo MG, Yang D, Mil- ler TE, Feldman J, Hauser BM, Caradonna TM, Clayton KL, Nitido AD, Murali MR, Alter G, Charles RC, Dighe A, Bran- da JA, Lennerz JK, Lingwood D, Schmidt AG, Iafrate AJ, Balazs AB. COVID-19-neutralizing antibodies predict disease severity and survival. Cell 2021; 184: 476-488.e11.
23. Sette A, Crotty S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 2021; 184: 861-880.
24. Ng KW, Faulkner N, Cornish GH, Rosa A, Harvey R, Hussain S, Ulferts R, Earl C, Wrobel AG, Benton DJ, Rou- stan C, Bolland W, Thompson R, Agua-Doce A, Hobson P, Heaney J, Rickman H, Paraskevopoulou S, Houlihan CF, Thomson K, Sanchez E, Shin GY, Spyer MJ, Joshi D, O’Reilly N, Walker PA, Kjaer S, Riddell A, Moore C, Jebson BR, Wilkinson M, Marshall LR, Rosser EC, Radziszewska A, Peckham H, Ciurtin C, Wedderburn LR, Beale R, Swanton C, Gandhi S, Stockinger B, McCauley J, Gamblin SJ, McCoy LE, Cherepanov P, Nastouli E, Kassiotis G. Preexisting and de novo humoral immunity to SARS-CoV-2 in humans. Science 2020; 370: 1339-1343.
25. Nguyen-Contant P, Embong AK, Kanagaiah P, Chaves FA, Yang H, Branche AR, Topham DJ, Sangster MY. S Protein-reactive IGG and memory B cell production after human SARS-CoV-2 infection includes broad reactivity to the S2 subunit. mBio 2020; 11: e01991-20.
26. Varona JF, Madurga R, Peñalver F, Abarca E, Almirall C, Cruz M, Ramos E, Castellano-Vazquez JM. Kinetics of anti-SARS-CoV-2 antibodies over time. Results of 10 month follow up in over 300 seropositive health care workers. Eur J Intern Med 2021; 89: 97-103.
27. Liu G, Rusling JF. COVID-19 antibody tests and their limitations. ACS Sens 2021; 6: 593-612.
28. Ng KW, Faulkner N, Cornish GH, Rosa A, Harvey R, Hussain S, Ulferts R, Earl C, Wrobel AG, Benton DJ, Rou- stan C, Bolland W, Thompson R, Agua-Doce A, Hobson P, Heaney J, Rickman H, Paraskevopoulou S, Houlihan CF, Thomson K, Sanchez E, Shin GY, Spyer MJ, Joshi D, O’Reilly N, Walker PA, Kjaer S, Riddell A, Moore C, Jebson BR, Wilkinson M, Marshall LR, Rosser EC, Radziszewska A, Peckham H, Ciurtin C, Wedderburn LR, Beale R, Swanton C, Gandhi S, Stockinger B, McCauley J, Gamblin SJ, McCoy LE, Cherepanov P, Nastouli E, Kassiotis G. Preexisting and de novo humoral immunity to SARS-CoV-2 in humans. Science 2020; 370: 1339-1343.
29. Hicks J, Klumpp-Thomas C, Kalish H, Shunmugavel A, Mehalko J, Denson JP, Snead KR, Drew M, Corbett KS, Graham BS, Hall MD, Memoli MJ, Esposito D, Sadtler K. Serologic cross-reactivity of SARS-CoV-2 with endemic and seasonal betacoronaviruses. J Clin Immunol 2021; 41: 906-913.
30. Lv H, Wu N, Tsang OTY, Yuan M, Perera RAPM, Leung WS, So RTY, Chan JMC, Yip GK, Chik TSH, Wang Y, Choi CYC, Lin Y, Ng WW, Zhao J, Poon LLM, Peiris JSM, Wilson IA, Mok CKP. Cross-reactive antibody response between SARS-CoV-2 and SARS-CoV infections. Cell Rep 2020; 31: 107725.
31. Al-Alawi S, Al-Hinai H, Al-Kindi N, Al-Rashidi M, Al-Kindi H, Al-Shukri I, Al-Rashdi A, Jose S, Al-Jardani A. Evaluation of four rapid antigen tests for detection of SARS-CoV-2 virus. Oman Med J 2021; 36: e273.
32. Hsieh WY, Lin CH, Lin TC, Lin CH, Chang HF, Tsai CH, Wu HT, Lin CS. Development and efficacy of lateral flow point-of-care testing devices for rapid and mass COVID-19 diagnosis by the detections of SARS-CoV-2 antigen and anti-SARS-CoV-2 antibodies. Diagnostics 2021; 11: 1760.
33. Kopel J, Goyal H, Perisetti A. Antibody tests for COVID-19. Proc Bayl Univ Med Cent 2021; 34: 63-72.
34. Mouliou DS, Gourgoulianis KI. False-positive and false-negative COVID-19 cases: respiratory prevention and management strategies, vaccination, and further perspectives. Expert Rev Respir Med 2021; 15: 993-1002.
35. Jalkanen P, Kolehmainen P, Häkkinen HK, Huttunen M, Tähtinen PA, Lundberg R, Maljanen S, Reinholm A, Tauriainen S, Pakkanen SH, Levonen I, Nousiainen A, Miller T, Välimaa H, Ivaska L, Pasternack A, Naves R, Ritvos O, Österlund P, Kuivanen S, Smura T, Hepojoki J, Vapalahti O, Lempainen J, Kakkola L, Kantele A, Julkunen I. COVID-19 mRNA vaccine induced antibody responses against three SARS-CoV-2 variants. Nat Commun 2021; 12: 3991.
36. Martínez-Baz I, Miqueleiz A, Casado I, Navascués A, Trobajo-Sanmartín C, Burgui C, Guevara M, Ezpeleta C, Castilla J; Working Group for the Study of COVID-19 in Navarra. Effectiveness of COVID-19 vaccines in preventing SARS-CoV-2 infection and hospitalisation, Navarre, Spain, January to April 2021. Euro Surveill 2021; 26: 2100438.
37. Ibarrondo FJ, Hofmann C, Fulcher JA, Goodman-Meza D, Mu W, Hausner MA, Ali A, Balamurugan A, Taus E, Elliott J, Krogstad P, Tobin NH, Ferbas KG, Kitchen SG, Aldrovandi GM, Rimoin AW, Yang OO. Primary, recall, and decay kinetics of SARS-CoV-2 vaccine antibody responses. ACS Nano 2021; 15: 11180-11191.
38. Tada T, Zhou H, Dcosta BM, Samanovic MI, Dcosta BM, Cornelius A, Mulligan MJ, Landau NR. Comparison of neutralizing antibody titers elicited by mRNA and adenoviral vector vaccine against SARS-CoV-2 variants. bioRxiv 2021: 2021.07.19.452771.
39. Nordström P, Ballin M, Nordström A. Effectiveness of heterologous ChAdOx1 nCoV-19 and mRNA prime-boost vaccination against symptomatic COVID-19 infection in Sweden: a nationwide cohort study. Lancet Reg Health Eur 2021; 11: 100249.
40. Barros-Martins J, Hammerschmidt SI, Cossmann A, Odak I, Stankov MV, Ramos GM, Dopfer-Jablonka A, Heidemann A, Ritter C, Friedrichsen M, Schultze-Florey C, Ravens I, Willenzon S, Bubke A, Ristenpart J, Janssen A, Ssebyatika G, Bernhardt G, Münch J, Hoffmann M, Pöhlmann S, Krey T, Bošnjak B, Förster R, Behrens GMN. Immune responses against SARS-CoV-2 variants after heterologous and homologous ChAdOx1 NCoV-19/BNT162b2 vaccination. Nat Med 2021; 27: 1525-1529.
41. Muik A, Lui BG, Wallisch AK, Bacher M, Mühl J, Reinholz J, Ozhelvaci O, Beckmann N, de la Caridad Güimil Gar- cia R, Poran A, Shpyro S, Finlayson A, Cai H, Yang Q, Swan- son KA, Türeci Ö, Şahin U. Neutralization of SARS-CoV-2 omicron by BNT162b2 mRNA vaccine-elicited human sera. Science 2022; 375: 678-680.
42. Khoury DS, Cromer D, Reynaldi A, Schlub TE, Whea- tley AK, Juno JA, Subbarao K, Kent SJ, Triccas JA, Davenport MP. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med 2021; 27: 1205-1211.
43. Modenese A, Paduano S, Bellucci R, Marchetti S, Bruno F, Grazioli P, Vivoli R, Gobba F, Bargellini A. Investigation of possible factors influencing the neutralizing anti-SARS-CoV-2 antibody titer after six months from the second vaccination dose in a sample of italian nursing home personnel. Antibodies (Basel) 2022; 11: 59.
44. Hurt AC, Wheatley AK. Neutralizing antibody therapeutics for Covid-19. Viruses 2021; 13: 628.
45. Platzer B, Stout M, Fiebiger E. Antigen cross-presentation of immune complexes. Front Immunol 2014; 5: 140.
46. Mueller AL, McNamara MS, Sinclair DA. Why does COVID-19 disproportionately affect older people? Aging (Albany NY) 2020; 12: 9959-9981.
47. Fung M, Babik JM. COVID-19 in immunocompromised hosts: what we know so far. Clin Infect Dis 2021; 72: 340-350.
48. COVID-19 Host Genetics Initiative. Mapping the human genetic architecture of COVID-19. Nature 2021; 600: 472-477.
49. Jayawardena R, Sooriyaarachchi P, Chourdakis M, Jeewandara C, Ranasinghe P. Enhancing immunity in viral infections, with special emphasis on COVID-19: a review. Diabetes Metab Syndr 2020; 14: 367-382.
50. Williamson EJ, Walker AJ, Bhaskaran K, Bacon S, Bates C, Morton CE, Curtis HJ, Mehrkar A, Evans D, Inglesby P, Cockburn J, McDonald HI, MacKenna B, Tomlinson L, Douglas IJ, Rentsch CT, Mathur R, Wong AYS, Grieve R, Harrison D, Forbes H, Schultze A, Croker R, Parry J, He- ster F, Harper S, Perera R, Evans SJW, Smeeth L, Goldacre B. Factors associated with COVID-19-related death using OpenSAFELY. Nature 2020; 584: 430-436.
51. Krammer F, Srivastava K, Alshammary H, Amoako AA, Awawda MH, Beach KF, Bermúdez-González MC, Bie- lak DA, Carreño JM, Chernet RL, Eaker LQ, Ferreri ED, Floda D, Gleason CR, Hamburger JZ, Jiang K, Kleiner G, Jurczyszak D, Matthews JC, Mendez WA, Nabeel I, Mulder LCF, Raskin AJ, Russo Kayla T, Salimbangon ABT, Saksena M, Shin AS, Singh G, Sominsky LA, Stadlbauer D, Wajnberg A, Simon V. Antibody responses in seropositive persons after a single dose of SARS-CoV-2 mRNA vaccine. N Engl J Med 2021; 384: 1372-1374.
52. BMJ. COVID-19: The BMJ’s Coronavirus Hub [Internet]. BMJ. [cited 2023 Apr 25]. https://www.bmj.com/content/380/bmj.p264
53. MAGICapp. National COVID-19 Clinical Evidence Taskforce [Internet]. Australian Government Department of Health. [cited 2023 Apr 25]. https://app.magicapp.org/#/guideline/6914/section/121234
54. Zhou P, Song G, Liu H, Yuan M, He WT, Beutler N, Zhu X, Tse LV, Martinez D, Schäfer A, Anzanello F, Yong P, Peng L, Dueker K, Musharrafieh R, Callaghan S, Capozzola T, Limbo O, Parren M, Garcia E, Rawlings SA, Smith DM, Nemazee D, Jardine JG, Safonova Y, Briney B, Rogers TF, Wilson IA, Baric RS, Gralinski LE, Burton DR, Andrabi R. Broadly neutralizing anti-S2 antibodies protect against all three human betacoronaviruses that cause deadly disease. Immunity 2023; 56: 669-686.
55. Kim DM, Kim HS, Seo JW, Lee J, Park U, Ha NY, Koh J, Park H, Lee JW, Ro HJ, Yun NR, Kim DY, Yoon SH, Na YS, Moon DS, Lim SC, Kim CM, Jeon K, Kang JG, Jang NY, Jeong H, Kim J, Cheon S, Sohn KM, Moon JY, Kym S, Han SR, Lee MS, Kim HJ, Park WY, Choi JY, Shin HW, Kim HY, Cho CH, Jeon YK, Kim YS, Cho NH. Enhanced eosinophil-mediated inflammation associated with antibody and complement-dependent pneumonic insults in critical COVID-19. Cell Rep 2021; 37: 109798.
56. Ankerhold J, Giese S, Kolb P, Maul-Pavicic A, Voll RE, Göppert N, Ciminski K, Kreutz C, Lother A, Salzer U, Bildl W, Welsink T Morgenthaler NG, Grawitz AB, Huzly D, Schwemmle M, Hengel H, Falcone V. Circulating multimeric immune complexes drive immunopathology in COVID-19. bioRxiv 2021 Jun 25. doi: 10.1101/2021.06.25.449893.
57. Bye AP, Hoepel W, Mitchell JL, Jégouic S, Loureiro S, Sage T, Vidarsson G, Nouta J, Wuhrer M, de Taeye S, van Gils M, Kriek N, Cooper N, Jones I, den Dunnen J, Gibbins JM. Aberrant glycosylation of anti-SARS-CoV-2 spike IgG is a prothrombotic stimulus for platelets. Blood 2021; 138: 1481-1489.
58. Nazy I, Jevtic SD, Moore JC, Huynh A, Smith JW, Kel- ton JG, Arnold DM. Platelet activating immune complexes identified in COVID-19 associated coagulopathy. medRxiv. 2020 Nov 4. doi: 10.1101/2020.11.04.20226076.
59. Bastard P, Rosen LB, Zhang Q, Michailidis E, Hof- fmann HH, Zhang Y, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science 2020; 370: eabd4585. doi: 10.1126/science.abd4585.
60. Pfister F, Vonbrunn E, Ries T, Jäck HM, Überla K, Lochnit G, Sheriff A, Herrmann M, Büttner-Herold M, Amann K, Daniel C. Complement activation in kidneys of patients with COVID-19. Front Immunol 2020; 11: 594849.
Copyright: © 2023 Jan Kochanowski University in Kielce This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
© 2024 Termedia Sp. z o.o.
Developed by Bentus.