650
Views
1
CrossRef citations to date
0
Altmetric
Commentary

Antibodies: what makes us stronger

Pages 3551-3553 | Received 26 Mar 2021, Accepted 06 May 2021, Published online: 25 May 2021

ABSTRACT

Neutralizing antibodies are the basis of almost all approved prophylactic vaccines and the foundation of effective protection from pathogens, including the recently emerging SARS Coronavirus 2 (SARS-CoV-2). However, the contribution of antibodies to protection and to the course of the disease during first-time exposure to a pathogen is unknown. We analyzed the antibodies and B cell responses in severe and mild COVID-19 patients. Despite our primary assumption that high antibody titers contribute to a mild disease, we found that severe COVID-19 illness, and not mild infection, correlates with strong anti-viral antibody and memory B cell responses. This phenomenon was also demonstrated for anti-Mycobacterium tuberculosis inhibiting antibodies that we recently isolated from an actively infected Tuberculosis-sick donor. This correlation between disease severity and antibody responses can be explained by the fact that high viral loads drive B cell stimulation and generation of high-affinity antibodies that will be protective upon future encounter with the particular pathogen.

Our immune response is comprised of two main arms; the first is the innate immunity arm, which functions as an immediate line of response to pathogens and its components include mainly activated lymphocytes and epithelial cells, as well as soluble factors, such as proteins of the complement pathway. The second arm of our immune system is the adaptive immunity, which is separated into cellular and humoral immunity, represented by T cells and B cells, respectively. B cell immunity is responsible for the generation of antibodies and it is an important component of effective protection from pathogens. When infection occurs, naïve B cells are stimulated, and through a unique process called “affinity maturation,” differentiate into memory B cells and antibody-secreting plasma cells.Citation1 In some infections, plasma cells secreting pathogen-specific antibodies are detected as early as on day 4 post infection, peaking around days 7 post infection.Citation2 The pathogen-specific antibodies can directly bind to the infectious agent or bacteria-specific toxin and sometimes, this binding event itself blocks the pathogen and subsequently prevents the infection or blocks the biological activity of the toxin. These types of antibodies are commonly termed “neutralizing antibodies,” and they have been defined as one of the most important factors in protection during infections and the basis of all approved vaccines.Citation3 The importance of antibodies in protection from pathogenic infections is also demonstrated by the fact that B cell immunodeficient individuals who are unable to mount IgG responses have recurrent and increased susceptibility to infections.Citation4

While the role of preexisting pathogen-specific antibodies in protection from infections has been set in stone, the role of pathogen-specific antibodies in protection during first-time encounter with the corresponding pathogen, is not yet clear. During first-time exposure to a pathogen, the B cell response is maturing in parallel with the developing infection. Therefore, one might ask what is the correlation between asymptomatic first-time infection and the production of highly effective and robust neutralizing antibodies, and is failure to produce such an effective antibody response during the first-time encounter with the pathogen result in greater morbidity? The emergence of the COVID-19 pandemic provided a unique opportunity to systematically address this question by studying the relationships between a new virus previously unknown to the immune system, a new disease, and the antibodies co-evolving against it.

Correlation between COVID-19 disease severity and serum IgG

Infection with the recently emerging SARS Coronavirus 2 (SARS-CoV-2) results in severe disease manifestations in approximately 10% of the virologically confirmed infections, leading to death in 1–4% of all confirmed infection.Citation5,Citation6 Nonetheless, the vast majority (according to most reports between 60% and 75% of the infectees) demonstrate a mild disease that clears on its own, whereas unundefined portion (according to some reports between 20% and 40% of the infectionsCitation6,Citation7) is asymptomatic. Given the wide range of possible clinical outcomes, and the fact that antibody responses against SARS-CoV-2 have been reported very early in the course of the pandemic,Citation8 we hypothesized that mild asymptomatic infection correlates with high levels of antibodies that hold SARS-CoV-2 back, thus preventing viral spread and subsequent tissue damage. With this assumption in mind, our group initiated a search for highly potent anti-SARS-CoV-2 neutralizing antibodies in mild SARS-CoV-2 infectees that will serve as candidates for therapeutics, and provide insights for vaccine targets. However, analysis of serological and B cell responses in convalescent mildly and severely sick patients by our group,Citation9 as well as by othersCitation10–12 revealed a reversed picture, thus refuting our primary postulation. Our in-depth analyses of B cells showed that in the case of COVID-19, it is a severe disease that is associated with both higher titers and better antibody responses, which are directed against numerous nonoverlapping epitopes on the virus.Citation9 In contrast to our initial assumption, mild infection resulted in significantly lower titers of inhibiting antibodies (p < .0001),Citation9 as assayed by the ability of the serum to inhibit SARS-CoV-2 Spike receptor-binding domain (RBD) and ACE2 interaction. RBD is a major neutralizing component on the SARS-CoV-2 and the majority of neutralizing anti-SARS-CoV-2 antibodies are directed against this component.Citation13 We also found greater B cell clonal expansion and higher frequencies of anti-SARS-CoV-2 Spike receptor-binding domain (RBD) memory B cells, in severe convalescent SARS-CoV-2 donors compared to asymptomatic SARS-CoV-2-infected individuals.Citation9 The severe donors we analyzed developed multi-clonal neutralizing anti-SARS-CoV-2 antibodies directed against different sites on the SARS-CoV-2 Spike RBD.Citation9 When combined, the neutralizing antibodies from one of the severe donors we analyzed, CoV02, were capable of suppressing the virus for several days in culture, more effectively than the individual effect of each single antibody, suggesting that the antibody response in this donor was not only neutralizing but also synergistic.Citation9

In fact, the correlation between severe disease caused by a high pathogenic burden and effective antibody neutralization is not limited to COVID-19. In HIV-1 infection, anti-HIV-1 broadly neutralizing antibodies were isolated from individuals with high viral loads.Citation14 Several studies showed that viral stimulation is needed to drive the evolution of neutralizing responses, occurring in ‘waves’ that follow the preceding ‘waves’ of viremia in donors with chronic HIV-1 infection.Citation15,Citation16 A unique subset of HIV-1-infected individuals, elite HIV-1 controllers, who are able to maintain low levels of viremia without the use of antiviral drugs, are the source of several of the most potent anti-HIV-1 antibodies.Citation17–19 However, viral control in these individuals was found to be largely due to specific alleles of the human leukocyte antigen (HLA) types, resulting in highly effective CD8 + T cells response.Citation20 In these cases, HIV-1 is kept under control mainly by the T cell arm of the immune system; however, the fact that the virus is still chronically present (even if viremia is low) is sufficient to stimulate and drive the B cell arm. This condition enables the generation of extremely broad cross-clade neutralizing antibodies that sometimes coexist with the viral variants.Citation21

Inhibiting antibody responses in Mycobacterium tuberculosis actively infected donors

While it is becoming more and more appreciated, the role of antibodies in some bacterial infections, such as Mycobacterium tuberculosis (Mtb) is still controversial.Citation22,Citation23 Health-care workers repeatedly exposed to sick patients exhibited polyclonal serum antibodies that when passively transferred to mice, protected them from the Mtb challenge,Citation24 indicating that anti-bacterial inhibiting antibodies are produced upon exposure. Antibodies against Mtb lipopolysaccharide residues were shown to provide a certain degree of protection,Citation25 however the main protein-based targets for neutralizing antibodies remain to be largely unknown. This presents one of the major challenges in the field of anti-bacterial antibodies – the vast complexity of the bacteria and the high abundance of proteins compared to viruses that usually encode only a small number of structural proteins. We recently used a recombinant phosphate transporter protein (PstS1) to isolate protective anti-Mtb monoclonal antibodies from a symptomatic, actively infected patient who was hospitalized due to severe tuberculosis disease and treated until recovery.Citation26 The two antibodies, p4-36 and p4-163, showed 50% protection both in vitro and in a mouse model, when administered to BALB/c mice prior to infection.Citation26 Interestingly, affinity maturation was crucial for the activity of both of these antibodies as reversion of their somatic hypermutations canceled their activity.Citation26 This emphasizes the fact that while the donor himself was sick, B cell responses and affinity maturation were ongoing, resulting in the production of high-affinity antibodies. Although such antibodies did not reduce Tuberculosis disease loads in the donor during the time of their generation, they might be protective during future bacterial encounters. Monoclonal antibodies, such as p4-36 and p4-163 can be evaluated as potential biological therapeutics to serve as an adjunctive treatment of antibiotic-resistant strains, as well as provide important leads for vaccine design.

The correlation between the severity of the disease during first-time exposure and the quality of the produced antibodies fits Friedrich Nietzsche’s aphorism, “what does not kill me makes me stronger” – in this case, meaning that the high pathogenic loads ultimately push the B cell responses forward to generate an effective germinal center reaction, and subsequently produce an effective and diverse polyclonal response. In light of this, in the pursuit for the next effective neutralizing antibody, not only individuals who had mild disease should be considered, but one should also consider looking into the antibodies developed by donors who recovered from a severe illness, whose antibodies might be very effective.

Vaccination makes perfect

While the lack of symptoms following first-timeinfection may not be antibody-mediated,B cells and antibodies are the main mediators of protectionduring second-timeinfections. The concept of vaccination is based on the notion that the B cell community needs to be trained on the most relevant targets while depositing long-lastingpolyclonal and effective B cell memory. For this reason, vaccination can be even more effective than natural infection by providing an improved and targeted immune response compared to immune response to infection. Accordingly, it has been shown that while the B cell response following seasonal influenza vaccination is highly effective in neutralizing the virus, a significant fraction of the antibodies produced following influenza infection are based on recall of less effective memory B cells.Citation27 Similarly, in the case of COVID-19, anti-SARS-CoV-2 antibody responses following vaccination were shown to be more robust and effective compared to those produced in response to an infection.Citation28 Similar phenomena, i.e., that antibody response to a vaccine is advantageous to the response to natural infection, were reported for HPV.Citation29 In conclusion, the revolution of vaccines enables us to train B cells by direct and sufficient antigenic stimulation, without pathogen-induced disease, and circumventing the need to “survive” the first encounter with the virus or bacteria, while still gaining effective protection.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Acknowledgments

The author thanks Jonathan M. Gershoni for helpful discussions and Anna Roitburd-Berman for help with scientific editing. The author is supported by The Campbell Foundation for HIV-1/AIDS Research and by ISF grants #1422/18 and #3711/20.

References

  • Victora GD, Nussenzweig MC. Germinal centers. Annu Rev Immunol. 2012;30:429–57. doi:10.1146/annurev-immunol-020711-075032.
  • Carter MJ, Mitchell RM, Meyer Sauteur PM, Kelly DF, Truck J. The antibody-secreting cell response to infection: kinetics and clinical applications. Front Immunol. 2017;8:630. doi:10.3389/fimmu.2017.00630.
  • Crowe JE Jr. Principles of broad and potent antiviral human antibodies: insights for vaccine design. Cell Host Microbe. 2017;22:193–206. doi:10.1016/j.chom.2017.07.013.
  • Fried AJ, Bonilla FA. Pathogenesis, diagnosis, and management of primary antibody deficiencies and infections. Clin Microbiol Rev. 2009;22:396–414. doi:10.1128/CMR.00001-09.
  • Brehm TT, Van Der Meirschen M, Hennigs A, Roedl K, Jarczak D, Wichmann D, Frings D, Nierhaus A, Oqueka T, Fiedler W, et al. Comparison of clinical characteristics and disease outcome of COVID-19 and seasonal influenza. Sci Rep. 2021;11:5803. doi:10.1038/s41598-021-85081-0.
  • Wang J, Zheng X, Chen J. Clinical progression and outcomes of 260 patients with severe COVID-19: an observational study. Sci Rep. 2021;11:3166. doi:10.1038/s41598-021-82943-5.
  • Buitrago-Garcia D, Egli-Gany D, Counotte MJ, Hossmann S, Imeri H, Ipekci AM, Salanti G, Low N. Occurrence and transmission potential of asymptomatic and presymptomatic SARS-CoV-2 infections: a living systematic review and meta-analysis. PLoS Med. 2020;17:e1003346. doi:10.1371/journal.pmed.1003346.
  • Amanat F, Denolly S, Vogrig M, Boson B, Siret E, Rigaill J, Pillet S, Grattard F, Gonzalo S, Verhoeven P, et al. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat Med. 2020;26:1033–36. doi:10.1038/s41591-020-0913-5.
  • Mor M, Werbner M, Alter J, Safra M, Chomsky E, Lee JC, Hada-Neeman S, Polonsky K, Nowell CJ, Clark AE, et al. Multi-clonal SARS-CoV-2 neutralization by antibodies isolated from severe COVID-19 convalescent donors. PLoS Pathog. 2021;17:e1009165. doi:10.1371/journal.ppat.1009165.
  • Hansen CB, Jarlhelt I, Pérez-Alós L, Hummelshøj Landsy L, Loftager M, Rosbjerg A, Helgstrand C, Bjelke JR, Egebjerg T, Jardine JG, et al. SARS-CoV-2 antibody responses are correlated to disease severity in COVID-19 convalescent individuals. J Immunol. 2021;206:109–17. doi:10.4049/jimmunol.2000898.
  • Marklund E, Leach S, Axelsson H, Nyström K, Norder H, Bemark M, Angeletti D, Lundgren A, Nilsson S, Andersson L-M, et al. Serum-IgG responses to SARS-CoV-2 after mild and severe COVID-19 infection and analysis of IgG non-responders. PLoS One. 2020;15:e0241104. doi:10.1371/journal.pone.0241104.
  • Robbiani DF, Gaebler C, Muecksch F, Lorenzi JCC, Wang Z, Cho A, Agudelo M, Barnes CO, Gazumyan A, Finkin S, et al. Convergent antibody responses to SARS-CoV-2 in convalescent individuals. Nature. 2020;584:437–42. doi:10.1038/s41586-020-2456-9.
  • Huo J, Le Bas A, Ruza RR, Duyvesteyn HM, Mikolajek H, Malinauskas T, Tan TK, Rijal P, Dumoux M, Ward PN, et al. Neutralizing nanobodies bind SARS-CoV-2 spike RBD and block interaction with ACE2. Nat Struct Mol Biol. 2020. doi:10.1038/s41594-020-0469-6.
  • Freund NT, Horwitz JA, Nogueira L, Sievers SA, Scharf L, Scheid JF, Gazumyan A, Liu C, Velinzon K, Goldenthal A, et al. A new glycan-dependent CD4-binding site neutralizing antibody exerts pressure on HIV-1 in vivo. PLoS Pathog. 2015;11:e1005238. doi:10.1371/journal.ppat.1005238.
  • Liao HX, Lynch R, Zhou T, Gao F, Alam SM, Boyd SD, Fire AZ, Roskin KM, Schramm CA, Zhang Z, et al. Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature. 2013;496:469–76. doi:10.1038/nature12053.
  • Moore PL, Gray ES, Wibmer CK, Bhiman JN, Nonyane M, Sheward DJ, Hermanus T, Bajimaya S, Tumba NL, Abrahams M-R, et al. Evolution of an HIV glycan-dependent broadly neutralizing antibody epitope through immune escape. Nat Med. 2012;18:1688–92. doi:10.1038/nm.2985.
  • Mouquet H, Scharf L, Euler Z, Liu Y, Eden C, Scheid JF, Halper-Stromberg A, Gnanapragasam PNP, Spencer DIR, Seaman MS, et al. Complex-type N-glycan recognition by potent broadly neutralizing HIV antibodies. Proc Natl Acad Sci U S A. 2012;109:E3268–3277. doi:10.1073/pnas.1217207109.
  • Scheid JF, Mouquet H, Ueberheide B, Diskin R, Klein F, Oliveira TYK, Pietzsch J, Fenyo D, Abadir A, Velinzon K, et al. Sequence and structural convergence of broad and potent HIV antibodies that mimic CD4 binding. Science. 2011;333:1633–37. doi:10.1126/science.1207227.
  • Zhou T, Georgiev I, Wu X, Yang Z-Y, Dai K, Finzi A, Do Kwon Y, Scheid JF, Shi W, Xu L, et al. Structural basis for broad and potent neutralization of HIV-1 by antibody VRC01. Science. 2010;329:811–17. doi:10.1126/science.1192819.
  • Miura T, Brockman MA, Schneidewind A, Lobritz M, Pereyra F, Rathod A, Block BL, Brumme ZL, Brumme CJ, Baker B, et al. HLA-B57/B*5801 human immunodeficiency virus type 1 elite controllers select for rare gag variants associated with reduced viral replication capacity and strong cytotoxic T-lymphocyte [corrected] recognition. J Virol. 2009;83:2743–55. doi:10.1128/JVI.02265-08.
  • Freund NT, Wang H, Scharf L, Nogueira L, Horwitz JA, Bar-On Y, Golijanin J, Sievers SA, Sok D, Cai H, et al. Coexistence of potent HIV-1 broadly neutralizing antibodies and antibody-sensitive viruses in a viremic controller. Sci Transl Med. 2017;9:eaal2144. doi:10.1126/scitranslmed.aal2144.
  • Li H, Javid B. Antibodies and tuberculosis: finally coming of age? Nat Rev Immunol. 2018;18:591–96. doi:10.1038/s41577-018-0028-0.
  • Casadevall A. Antibodies to Mycobacterium tuberculosis. N Engl J Med. 2017;376:283–85. doi:10.1056/NEJMcibr1613268.
  • Li H, Wang -X-X, Wang B, Fu L, Liu G, Lu Y, Cao M, Huang H, Javid B. Latently and uninfected healthcare workers exposed to TB make protective antibodies against Mycobacterium tuberculosis. Proc Natl Acad Sci U S A. 2017;114:5023–28. doi:10.1073/pnas.1611776114.
  • Correia-Neves M, Sundling C, Cooper A, Kallenius G. Lipoarabinomannan in active and passive protection against tuberculosis. Front Immunol. 2019;10:1968. doi:10.3389/fimmu.2019.01968.
  • Watson A, Li H, Ma B, Weiss R, Bendayan D, Abramovitz L, Ben-Shalom N, Mor M, Pinko E, Oz MB, et al. Human antibodies targeting a Mycobacterium transporter protein mediate protection against tuberculosis. Nat Commun. 2021;12:602. doi:10.1038/s41467-021-20930-0.
  • Dugan HL, Guthmiller JJ, Arevalo P, Huang M, Chen Y-Q, Neu KE, Henry C, Zheng N-Y, Lan LYL, Tepora ME, et al. Preexisting immunity shapes distinct antibody landscapes after influenza virus infection and vaccination in humans. Sci Transl Med. 2020;12:eabd3601. doi:10.1126/scitranslmed.abd3601.
  • Wang Z, Schmidt F, Weistblum Y, Muecksch F, Barnes CO, Finkin S, Schaefer-Babajew D, Cipolla M, Gaebler C, Lieberman JA, et al. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. Nature. 2021;592:616–22. doi:10.1038/s41586-021-03324-6.
  • Pattyn J, Van Keer S, Tjalma W, Matheeussen V, Van Damme P, Vorsters A. Infection and vaccine-induced HPV-specific antibodies in cervicovaginal secretions. A review of the literature. Papillomavirus Res. 2019;8:100185. doi:10.1016/j.pvr.2019.100185.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.