Next Article in Journal
Monkeypox Virus in Animals: Current Knowledge of Viral Transmission and Pathogenesis in Wild Animal Reservoirs and Captive Animal Models
Previous Article in Journal
Characterization of Anti-Poliovirus Compounds Isolated from Edible Plants
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

HLA Variation and SARS-CoV-2 Specific Antibody Response

by
Dawit Wolday
1,2,*,
Chun Yiu Jordan Fung
1,2,
Gregory Morgan
1,2,3,
Selina Casalino
1,2,
Erika Frangione
1,2,
Jennifer Taher
1,3 and
Jordan P. Lerner-Ellis
1,2,3,*
1
Pathology and Laboratory Medicine, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1Z5, Canada
2
Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON M5G 1Z5, Canada
3
Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1Z5, Canada
*
Authors to whom correspondence should be addressed.
Viruses 2023, 15(4), 906; https://doi.org/10.3390/v15040906
Submission received: 15 March 2023 / Revised: 29 March 2023 / Accepted: 29 March 2023 / Published: 31 March 2023
(This article belongs to the Section SARS-CoV-2 and COVID-19)

Abstract

:
Differences in SARS-CoV-2-specific immune responses have been observed between individuals following natural infection or vaccination. In addition to already known factors, such as age, sex, COVID-19 severity, comorbidity, vaccination status, hybrid immunity, and duration of infection, inter-individual variations in SARS-CoV-2 immune responses may, in part, be explained by structural differences brought about by genetic variation in the human leukocyte antigen (HLA) molecules responsible for the presentation of SARS-CoV-2 antigens to T effector cells. While dendritic cells present peptides with HLA class I molecules to CD8+ T cells to induce cytotoxic T lymphocyte responses (CTLs), they present peptides with HLA class II molecules to T follicular helper cells to induce B cell differentiation followed by memory B cell and plasma cell maturation. Plasma cells then produce SARS-CoV-2-specific antibodies. Here, we review published data linking HLA genetic variation or polymorphisms with differences in SARS-CoV-2-specific antibody responses. While there is evidence that heterogeneity in antibody response might be related to HLA variation, there are conflicting findings due in part to differences in study designs. We provide insight into why more research is needed in this area. Elucidating the genetic basis of variability in the SARS-CoV-2 immune response will help to optimize diagnostic tools and lead to the development of new vaccines and therapeutics against SARS-CoV-2 and other infectious diseases.

1. Introduction

SARS-CoV-2 infection leads to a diverse spectrum of clinical outcomes ranging from asymptomatic to critical clinical presentation and death. Established factors influencing these diverse clinical outcomes include age, sex, racial ancestry, comorbidities, coinfections, and SARS-CoV-2 variants [1,2,3,4,5,6,7]. Additionally, genetic factors may play a significant role in the pathogenesis of COVID-19-associated severity, reviewed in [8].
The human leukocyte antigen (HLA) system plays a crucial role in the host’s immune response during an encounter with an infectious agent [9]. The HLA system is further classified into HLA class I (HLA-A, HLA-B, and HLA-C) and HLA class II (HLA-DR, HLA-DQ, and HLA-DP). Whereas HLA class I is involved in the presentation of antigens in an endogenous pathway, HLA class II is engaged in an exogenous antigen presentation pathway [9]. HLA class I molecules are expressed on all nucleated cells and HLA class II molecules are expressed on specialized or professional antigen-presenting cells (APCs), such as dendritic cells, macrophages, and mature B cells. The HLA genes are located within the major histocompatibility complex (MHC). In humans, the MHC gene locus located in the short arm of chromosome six is among the most complex systems in humans. There are more than 15,000 genetic variations in the HLA class I and class II genes [9]; this, combined with the heterogeneity in antibody response and the variability of analytic methods have made it difficult to study. Nevertheless, among the genetic factors that determine the clinical outcome upon exposure to antigens, the HLA system plays a pivotal role in the immune response to various infections, such as viral hepatitis, dengue, HIV-1, Mycobacterium tuberculosis, and malaria, as well as small pox, rotavirus, measles-mumps-rubella, and influenza vaccines [10,11,12,13,14,15,16,17,18,19,20,21,22]. Likewise, emerging data show that polymorphisms in the HLA system may confer protection from or susceptibility to infection and severe disease in patients with SARS-CoV-2 infection, reviewed in [23,24]. In addition, the genetic makeup of an individual host may also modulate the response to SARS-CoV-2 vaccination [24,25]. Indeed, differences in SARS-CoV-2-specific immune responses have been observed between individuals following SARS-CoV-2 infection or vaccination [26,27,28,29], and HLA variation may contribute to such differences [24,25].
Understanding the underlying mechanisms of the relationship between HLA variation and SARS-CoV-2-specific antibody responses may provide an avenue for the development of novel therapeutic and preventive strategies for SARS-CoV-2, and consequently, the prevention of long-term sequelae of SARS-CoV-2 infection. In this article, we review the current evidence and discuss future prospects.

2. Host Immune Response to SARS-CoV-2: Innate and Adaptive Immunity

SARS-CoV-2 enters the respiratory tract’s airway epithelial cells via the angiotensin-converting enzyme 2 (ACE2)—the host receptor for the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 [30]. The cell surface-associated transmembrane serine protease (TMPRSS2) regulates the binding of the RBD to the ACE2 receptor that eventually triggers endocytosis of the virus, followed by the release of the viral mRNA into the host cells’ cytoplasm [31]. Within the cytoplasm, the virus hijacks the host cell machinery to initiate the replication and release of new viral particles. The release of damage-associated molecular patterns (DAMP) along with microorganism-associated molecular patterns (MAMPs) is followed by the host’s pattern-recognition receptors (PRRs) recognizing the neighboring airway cells, and the recruitment of a multitude of immune cells, including APCs, which present SARS-CoV-2 antigens during the generation of adaptive immune responses. Hence, the initial immune response characterized by the activation of innate immunity is followed by a virus-specific adaptive immune response.
During the generation of adaptive immune responses, the HLA molecules are involved in SARS-CoV-2 antigen presentation [32,33]. Following SARS-CoV-2 infection or vaccination, the spike antigen is taken up by antigen presenting cells, such as dendritic cells, macrophages, and B-cells, and processed into smaller peptides. As depicted in Figure 1, dendritic cells and alveolar macrophages present the peptides associated with HLA class I molecules to the T cell receptor (TCR) of cytotoxic CD8+ T cells (CTLs), leading to the death of SARS-CoV-2 infected cells [34,35,36]. On the other hand, dendritic cells present the peptides with HLA class II molecules to the TCR of naïve CD4+ helper (TH0) cells that differentiate into T follicular helper cells (TFH) that, in turn, induce B cell differentiation in the germinal center of draining lymph nodes that will eventually mature into memory B cells and plasma cells [37,38]. The plasma cells secrete SARS-CoV-2 specific neutralizing antibodies that block the interaction between the virus and the ACE-2 receptor [39,40]. In addition to inducing B cell differentiation, CD4+ T cells also induce the activation of CD8+ CTLs [41,42]. Differences in antibody responses have been observed based on the antigen target, including the spike protein and its RBD, and the nucleocapsid protein (NP), with the former correlating with viral neutralization [43,44]. Additionally, antibodies may play a protective role through the mechanisms involving antibody-mediated phagocytosis (AMP) or antibody-dependent cellular cytotoxicity (ADCC), by involving macrophages and natural killer cells, respectively [43,44]. Although immunoglobulin G (IgG) are relevant to durable humoral immune responses, immunoglobulin M (IgM) isotypes play a significant role during the acute phase of infection, and immunoglobulin A (IgA) antibodies are involved in the mucosal defense against SARS-CoV-2 [43,44]. The activation of SARS-CoV-2-specific immune cells leads to the death of infected cells, and the majority of patients subsequently clear the virus and recover [36]. In contrast, in those who develop severe disease, cytokines and chemokines continue to attract monocytes, macrophages, neutrophils, and T cells to the site of the infection, promoting further inflammation and the uncontrolled production of pro-inflammatory cytokines (also known as a cytokine storm), vascular endotheliitis, thrombosis, and angiogenesis [31,45,46].
One of the most peculiar characteristics of SARS-CoV-2 infection, in terms of the humoral antibody responses, is the extreme inter-individual variability. Although the exact underlying mechanism(s) have not been fully elucidated, the observed inter-individual variability in antibody responses has previously been associated with age [47,48], sex [48], comorbid conditions [48,49,50,51], smoking [47,48], COVID-19 severity [51,52], medications [49,50], viral lineage [53,54], SARS-CoV-2 vaccination type and dose [28,55,56], hybrid immunity [57,58], and time since SARS-CoV-2 infection or vaccination [59]. HLA alleles exhibit a high degree of variation [9]. Whether the differences observed in SARS-CoV-2 specific antibody responses between individuals are due to differences in the capability of the HLA molecules in presenting several peptides is not fully understood.

3. HLA Variation and Humoral Immunity in SARS-CoV-2

We conducted a literature search of the databases in PubMed and Google Scholar with the keywords “HLA” and “antibody response”, or “immunoglobulin response”, in individuals following “SARS-CoV-2 infection”, or “COVID-19”, and/or “vaccination”. The study cohort characteristics, including infection status, vaccination status, number of study participants and date, and antibody response-type determinations are summarized in Table 1. There was extreme heterogeneity in the study designs, sample sizes, and duration of follow-up [60,61,62,63,64,65,66,67,68,69]. In addition, the humoral immune responses were assessed by measuring antibody titers based on anti-RBD IgG isotypes in two-thirds of the studies [60,61,62,64,66,68], and three studies determined anti-Spike IgG isotype levels [61,63,65]. In another study, the antibody levels evaluated included anti-RBD IgA isotype and anti-NP total Ig levels [66]. Only three studies determined the levels of neutralizing antibodies [64,66,67]. Whereas high antibody responders were defined as those having the top 25th or 33rd percentile of the titer distribution [60,64,67], low antibody response was defined as having the lowest 5th or 33rd percentile of the titer distribution [65,68]. Other investigators compared differences in the median or mean titers between carriers and non-carriers [61,62,63,64,65,66,67]. While all the studies included MHC class II association with antibody responses, only five studied MHC class I association with antibody responses. Except for one study [61] which included children, all the remaining studies included adults only.
Though research on the association between HLA variants and humoral immune responses is emerging, the reports revealed conflicting results (Figure 2). Several recent studies provided evidence that specific HLA variation, namely DQA1*03:03, DQB1*06, DRB1*03:01, or DRB1*07:01, enhanced the serological response post vaccination [60,61,62,63,64]. In a study conducted in Japan involving 100 health care workers, vaccination with the BNT162b2 vaccine was followed by a significant anti-RBD IgG response in individuals with the DQA1*03:03:01 haplotype [60]. Interestingly, individuals who received two doses of the BNT162b2 vaccine and carry the DQB1*06:01 allele also showed protection against the decline of anti-RBD IgG titers [60]. A recent study by Mentzer and colleagues in the UK performed one of the most detailed HLA class II genomic analyses with the largest sample size (n = 2753) undertaken to date [61]. In this study, the investigators reported significantly higher levels of anti-RBD and/or anti-S IgG antibody responses in individuals carrying.
DQB1*06 alleles who received the ChAdOx1 one vaccine dose or were boosted with the same vaccine, or the BNT162b2 or mRNA-1273 vaccines. Interestingly, individuals carrying the DQB1*06 allele were less likely to exhibit breakthrough infections compared to non-carriers. Furthermore, memory B-cell responses in DQB1*06 carriers were increased following vaccination, and anti-RBD IgG production in a cohort of individuals who received a booster vaccination persisted for several months. In another study of 420 UK participants who received a single dose of the BNT162b2 vaccine post natural infection, individuals with the DRB1*03:01 allele demonstrated higher titers of anti-RBD IgG, although the antibody titer abated (average of 121 days) after a second dose of the same vaccine, despite sustained T cell responses against the spike protein [62]. Interestingly, individuals carrying the DRB1*13:02 allele in this study exhibited a greater susceptibility to symptomatic disease [62]. Similarly, a study conducted in Spain demonstrated high anti-Spike IgG titers among 87 individuals with the DRB1*07:01 allele and DRB1*07:01~DQA1*02:01~DQB1*02:02 haplotype 30 days after a second dose of the mRNA-1273 SARS-CoV-2 vaccine [63]. A recent study by Higuchi et al. of 87 Japanese patients with rheumatoid arthritis, who were vaccinated with the BNT162b2 vaccine, demonstrated that DRB1*12:01 allele carrier frequency was higher in those individuals with high anti-RBD and neutralizing antibody responders [64]. Additionally, allele carrier frequencies of DRB1*15:01 were higher in those individuals with high neutralizing antibody responses [64].
Other studies have documented the HLA variations that led to a reduced serological response post vaccination or post infection. Cocchiolo et al. studied 111 individuals without prior infection who received two BNT162b2 SARS-CoV-2 vaccine doses [65]. This study documented a weak anti-Spike IgG response that was less than 5% of the lowest community antibody response 14+ days after the second vaccine dose, in individuals carrying A*03:01, A*33:03, and B*58:01 alleles, and A*24:02~C*07:01~B*18:01~DRB1*11:04 haplotype. Another study by Fischer et al. investigated 119 COVID-19 convalescent adults, with a median follow-up of 250 days, who were unvaccinated [66]. In this study, individuals with the the B*35:01 and DRB1*01:01 alleles exhibited reduced titers of anti-RBD IgG and anti-NP total IgG. Likewise, Gutiérrez-Bautista et al. demonstrated that individuals carrying the allele DRB1*01:01 exhibited low anti-S antibody levels 30 days after a second dose of the mRNA-1273 vaccine [63]. Despite lower antibody levels, both alleles were also associated with a shorter duration of COVID-19 disease, suggesting the protective role of specific HLA variations. Notably, it is assumed that higher antibody levels are generally associated with protection. However, previous studies have documented that higher antibody titers can be associated with COVID-19 severity [43,44], although another study found that more severe illness was associated with higher ratios of anti-NP antibodies compared to anti-Spike/RBD antibodies [52]. Hence, whether the protective effects of B*35:01 and DRB1*01:01 alleles on COVID-19 clinical outcomes [66] is related to the generation of antibodies remains to be determined. Further, a low anti-Spike IgG titer was noted in the UK study in individuals carrying the DRB1*04:04 variant after a single BNT162b2 mRNA-based vaccination in patients with prior SARS-CoV-2 infection [62].
Contrary to the evidence presented above, there is literature that supports the lack of an association between antibody response and HLA variation. In the study conducted in Japan by Khor et al., no association was shown between anti-RBD IgG responses and polymorphisms in the MHC class I HLA locus-A*, -B*, -C*, DPA1*, DPB1*, DQA1*, DQB1*, and DRB1* [60]. Likewise, Astbury et al. showed no association between antibody production and several DRB1* alleles [62]. Notably, this study demonstrated no association between anti-RBD response and the DRB1*07:01 allele, although in a previous study, DRB1*07:01 was associated with high anti-S antibody responses [63]. Fisher et al. also demonstrated that there was no association between B*15:01 or DQB1*03:02 and anti-RBD responses, nor between B*35:01 or DRB1*01:01 and anti-RBD IgA and neutralizing antibodies after natural SARS-CoV-2 infection [66]. Another study undertaken in Austria found no association between neutralizing IgG antibodies and COVID-19 patients (n = 84) with A*20, B*35, C*14, DPB1*23, DQB1*05, and DRB1*13 polymorphisms [67]. A smaller study involving 56 patients conducted in Italy by Ragone et al. demonstrated no association between DRB1* or DQA1* and anti-RBD IgG titers in healthy individuals, measured two weeks to four months after two doses of BNT162b2 vaccination [68]. Another recent study conducted in Germany, in albeit a small number of COVID-19 patients (n = 49) who were either unvaccinated or received the BNT162b2 or ChAdOx1 vaccines, demonstrated that there was no association between A*01, A*02, A*24, B*44, C*04, C*07, DP*04, DQ*03, DQ*05, DQ*06, or DR*13 and anti-RBD IgG responses [69].

4. SARS-CoV-2 Peptide Epitope Binding Affinity and Antibody Response

HLA molecules can only present SARS-CoV-2 peptides that have binding epitopes compatible with its specific antigen binding cleft [29,30,31]. This leads to a mechanism whereby different HLA alleles present diverse peptides derived from the same SARS-CoV-2 antigen. Having certain alleles can make the SARS-CoV-2 antigenic presentation more efficient, leading to a better humoral immune response, both in terms of quantity and breadth (or affinity). Thus, the presence of HLA molecules capable of presenting several peptides will lead to a more efficient humoral immune response with a higher antibody titer. Antibodies that functionally neutralize correlate with protective capacity [36,43,44]. On the contrary, some unique HLA variants with the capacity to handle a very limited number of antigens are faced with the consequences of an inefficient humoral immune response. Thus, the differences in the capabilities of the HLA alleles in handling SARS-CoV-2 antigens may be the reason for the observed differences in anti-SARS-CoV-2-specific immune responses between individuals, the clinical severity of COVID-19, and vaccine efficacy.
The peptide-binding affinity of HLA class I alleles shows diverse characteristics. Notably, none of the HLA class I variants were associated with a positive SARS-CoV-2-specific antibody generation [60,65,66,67]. For example, A*02:06, A*03:01, A*11:01, A*24:02, B*35, B*35:01, B*52:01, B*58:01, C*03:03, C*07:02, and C*12:02 are associated with low or no antibody production, despite binding strongly with their respective peptide fragments (Figure 2) [70,71,72,73,74,75,76,77,78,79]. Additionally, A*02:06, B*35:01, and B*58:01 have been shown to be protective against SARS-CoV-2 infection [66,72,78]. On the contrary, A*03:01, A*11:01, A*24:02, B*52:01, and C*12:02 are all associated with an increased risk for severe COVID-19 and to susceptibility for infection with SARS-CoV-2 [72,73,76,80,81,82,83,84,85,86]. B*15:01 and C*01:02 are unable to present sufficient amounts of peptide epitope [66,87,88], and these alleles are also associated with severe COVID-19, COVID-19-related death, or an increased susceptibility for SARS-CoV-2 infection [66,75,88,89]. Whether the association of these HLA class I variants with adverse COVID-19 outcomes is attributed to their lack of antibody and/or efficient CD8+ CTL response deserves further study. Hence, the most plausible explanation for MHC class I roles as a protective or detrimental allele might be related to their ability to modulate the immune response in an antibody-independent manner.
With regards to the MHC class II, peptide-binding prediction analyses have revealed that DRB1*03:01, DRB1*07:01, and DRB1*12:01 are all associated with a significant increase in SARS-CoV-2-specific antibody responses following the administration of mRNA SARS-CoV-2 vaccines [62,63,64], and are also able to bind epitope peptides with a strong affinity [68,71,78,79]. Interestingly, these alleles are associated with protection against severe COVID-19 as well as reduced susceptibility to SARS-CoV-2 infection [66,72,78,81,85,89,90]. Hence, the protection against and/or reduced susceptibility to COVID-19 in individuals carrying these alleles could be attributed, at least in part, to the strong peptide binding affinity and the increase in antibody responses. Although several HLA class II variants, including DPA1*01:03, DPA1*02:01, DPB1*02:01, DPB1*04:02, DQB1*03:01, DRB1*01:01, DRB1*04:01, DRB1*04:02, DRB1*04:05, DRB1*09:01, DRB1*11:01, DRB1*11:04, DRB1*13:01, DRB1*13:02, and DRB1*15:01, are able to bind SARS-CoV-2 peptides with a strong affinity [71,79,85], there is no association between these alleles and antibody production [60,62]. On the contrary, the study by Higuchi et al. demonstrated that DRB1*15:01 is associated with increased anti-RBD neutralizing antibody responses [64], and exhibits a strong epitope binding affinity [68,79]. However, it is not associated with protection against severe COVID-19 [89]. DRB1*04:01 and DRB1*11:04 are both protective against severe COVID-19 and associated with a reduced susceptibility risk against SARS-CoV-2 infection [85,90,91]. Hence, the protective role of these alleles might be mediated by antibody-independent mechanisms, such as through CD4+ and/or CD8+ immune responses. DRB1*09:01, DRB1*11:01, DRB1*13:01, DRB1*13:02, and DRB1*15:01 are all associated with an increased risk of severe COVID-19 [62,89,90,92], and may also operate through alternative pathways. Hence, the effect of the above HLA variants might be attributed to their impact on T cell responses rather than on humoral immunity. This notion is supported by the study conducted by Astbury et al., who demonstrated that DRB1*15:01 carriers exhibited a significant increase in T-cell response, assessed using an IFN-Ɣ ELISpot assay, despite the absence of humoral immunity [62]. DRB1*01:01 and DRB1*04:04 exhibit a strong peptide binding affinity [71,79]. However, both these alleles are associated with a low or no association in antibody production [62,66]. In particular, DRB1*01:01 is associated with a shorter disease duration and protection against severe COVID-19 [66]. Hence, its protective effects might be related to its actions through antibody-independent mechanistic pathways involving CD8+ CTLs. Whereas one report revealed no association between DRB1*07:01 and the anti-RBD level [62], another study demonstrated that it is associated with increased anti-S levels [63], which might be attributed to its ability to bind peptide epitopes strongly [71,79]. In addition, DRB1*07:01 is associated with protection against severe COVID-19 as well as reduced susceptibility risk for SARS-CoV-2 infection [89,90].
Finally, DQB1*06 is associated with a significant increase in anti-RBD and/or Spike IgG responses in ChAdOx1 or BNT162b2 vaccinated individuals [61], and exhibits low peptide-binding affinity [90]. Although Mentzer et al. demonstrated the reduced hazard of breakthrough infection in vaccinated individuals carrying DQB1*06, this allele has been associated with an increased susceptibility for SARS-CoV-2 infection in a previous study [90]. As noted in an earlier section of this review (Figure 2), several HLA class II variants have been associated with low or weak antibody responses [60,62,66]. A SARS-CoV-2 peptide-binding prediction analysis demonstrated that DQB1*03:02, DRB1*01:01, and DRB1*08:01 alleles are unable to bind viral peptides with high affinity [65,67,91], have a weak or no antibody response [62,66], and are associated with an increased risk for severe COVID-19 and susceptibility for infection with SARS-CoV-2 [66,71,78,81,91,93]. Taken together, the association of DQB1*03:02, DRB1*01:01, and DRB1*08:01 alleles with low SARS-CoV-2 epitope-binding affinity may lead to low or no antibody production, and increase the risk of individuals for severe COVID-19 outcomes. Additionally, DRB1:01:01 can induce regulatory T cell responses that may inhibit downstream humoral immune response and antibody generation [94].

5. Conclusions and Future Prospects

The studies reviewed here reveal the inter-individual variations in antibody responses associated with diverse HLA polymorphisms [60,61,62,63,64,65,66,67,68,69]. There are several study design factors that lead to variable results within the literature, including differences in study cohort characteristics, the timeframe in which antibody levels were assessed, different instrumentation and cutoffs used for antibody assessment, and types of antibodies assessed (isotypes, antigen targets, and neutralizing ability) (Table 1). Small sample sizes and extreme allelic heterogeneity pose significant power limitations. The lack of standardization within SARS-CoV-2 antibody assays limits comparisons between studies. Further, differences in vaccine types, SARS-CoV-2 variants, genotyping methods, and algorithms for predicting HLA haplotype may also impact results [95,96]. For instance, a range of sequencing or genotyping technologies were used, including Ion GeneStudio [97], PacBio [98], Illumina, and Affymetrix [99,100,101]. The targeted next-generation sequencing panels and/or differences in the PCR techniques applied had different levels of resolution for HLA calling. An array of different algorithms were used to predict the HLA genotype, including NGSengine [99], AllType NGS [102], TypeStream Visual [103], Imputation, and PHASE [104,105,106], each with different technical performance parameters which also limit comparisons between studies.
Given that the CD4+ TFH TCR:MHC class II interaction is necessary for the generation of antibodies, any associations found between MHC class I and antibody titers [45,46] may imply a coincidental rather than a causal association. This notion is supported by the fact that in all the studies included in this review, HLA class I polymorphisms were not observed to have any effect on SARS-CoV-2-specific antibody responses [61,66,67,68]. On the other hand, in the event that future studies demonstrate an association between HLA class I and antibody responses, the findings might be the result of indirect regulatory pathways rather than a direct CD8 + TCR:MHC class I interaction. For instance, this notion is supported by the observation that HLA-B encodes a microRNA that regulates IgA production [107]. Indeed, the fact that some HLA class I alleles have no effect on antibody generation despite a strong peptide binding affinity implies that such alleles modulate the immune response in an antibody-independent manner through effects on CD8+ CTL responses.
Notably, two-thirds of the studies documented associations between HLA class II polymorphisms and antibody responses [60,61,62,63,64,65,66]. In addition, alternative immune-genetic pathways may be involved in the generation of antibodies. Indeed, a more recent report demonstrated that genetic variation in the 3′ regulatory region 1 (3′RR1) of the human immunoglobulin heavy chain locus has been associated with significant effects on SARS-CoV-2-specific antibody responses following BNT162b2 mRNA vaccination [108]. Whereas single nucleotide polymorphisms (SNPs) in rs373084296, rs7494441, rs12896746, rs12896897, and rs7144089 of the 3′RR1 of the human immunoglobulin heavy chain were associated with high levels of antibodies, SNPs in rs12896746, rs12896897, and rs7144089 were linked to low levels of antibodies (<10th percentile). Alternatively, immunogenicity demonstrated in response to SARS-CoV-2 may be ascribed to the generation of T-cell responses without sufficient humoral immune responses [62], or more probably due to the indirect pathways and impact by non-MHC-related regulatory genes [107,109]. Indeed, a more recent study demonstrated that non-MHC genes were associated with significant antibody production in Italian health care workers who received a second dose of the BNT162b2 mRNA-based or ChAdOx1 adenovirus-based SARS-CoV-2 vaccination [110]. The gene variants involved included TP53 (rs1042522), ABO (rs657152), APOE (rs7412/rs429358), ACE2 (rs2285666), HLA-A (rs2571381/rs2499), and CRP (rs2808635/rs876538). All these alleles were associated with significant increases in anti-spike IgG, as well as neutralizing antibodies, between two weeks and six months post vaccination.
In addition, recent genome-wide association studies (GWAS) have identified several SNPs, including the BCL11A (rs1123573) and TAC4 (rs77534576) genes, that are associated with COVID-19 severity [111]. These genes are involved in B-cell lymphopoiesis, and the role variants in these loci play in SARS-CoV-2-specific antibody responses deserves further investigation. Other variations or SNPs related to the host’s PRRs could also affect antibody responses. For example, PRRs such as the Toll-like receptor 7 (TLR7) are essential for immune cell activation and the connection to antimicrobial adaptive immunity [112,113]. Hence, individuals with unique loss-of-function (LoF) variants in TLR7 (rs189681811 and rs147244662) that are associated with COVID-19 severity have been linked with an abrogated production of IFNI and II [112]. Whether such early antiviral immune responses related to LoF variants in TLR7 also affect subsequent SARS-CoV-2-specific humoral immunity remains to be elucidated.
In addition, several authorities have demonstrated the emergence of SARS-CoV-2 variants with an enhanced capability to circumvent antibody responses [114,115,116,117,118]. These emerging SARS-CoV-2 variants may also have the potential to interact with HLA molecules with different binding affinity characteristics. Indeed, recent studies have revealed a dramatic loss of peptide-binding affinity associated with a mutation of the spike protein in individuals carrying A*02:01, B*07:02, DRB1*03:01, and DRB1*15:01 alleles [58,119,120,121,122]. Taken together, the emergence of new SARS-CoV-2 variants will pose a significant challenge to the development of effective immune responses and warrants further investigation.
In conclusion, the HLA loci are highly heterogeneous [9], and differences in study design, methodological approaches, and biological complexity have made this region difficult to study [95,96]. Thus, further studies using larger cohorts are needed to determine if there are associations between HLA loci and the SARS-CoV-2-specific immune response. It is expected that GWAS will provide further evidence about how genetic variation influences the differential antibody response after SARS-CoV-2 infection and/or vaccination. However, the molecular pathways involved remain to be elucidated. Replication studies intended to investigate the functional interactions of causal variants are underway by our group [123]. Ultimately, identifying the underlying immune-genetic mechanisms will pave the way for the optimization of new diagnostic modalities and the development of improved vaccines and therapeutic options against SARS-CoV-2 and other infectious diseases.

Author Contributions

D.W., J.T. and J.P.L.-E. conceived the idea, and D.W. drafted the review. D.W., C.Y.J.F., G.M., S.C., E.F., J.T. and J.P.L.-E. all contributed to revisions of the article and approved the submitted version. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Canadian Institutes of Health Research (Funding Reference Number VR4-172753).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Antibody-dependent cellular cytotoxicity (ADCC); antibody-mediated phagocytosis (AMP); antigen-presenting cells (APCs); binding antibody unit (BAU); Coronavirus disease 2019 (COVID-19); cytotoxic T lymphocytes (CTLs); genome-wide association studies (GWAS); damage-associated molecular patterns (DAMP); human leukocyte antigen (HLA); loss-of-function (LoF); major histocompatibility complex (MHC); microorganism-associated molecular patterns (MAMPs); nucleocapsid protein (NP); pattern-recognition receptors (PRRs); severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2); receptor-binding domain (RBD); single nucleotide polymorphisms (SNPs); spike antigen (S); T cell receptor (TCR); T follicular helper cells (TFH); regulatory T cells (Treg).

References

  1. O’Driscoll, M.; Dos Santos, G.R.; Wang, L.; Cummings, D.A.T.; Azman, A.S.; Paireau, J.; Fontanet, A.; Cauchemez, S.; Salje, H. Age-specific mortality and immunity patterns of SARS-CoV-2. Nature 2021, 590, 140–145. [Google Scholar] [CrossRef] [PubMed]
  2. Takahashi, T.; Ellingson, M.K.; Wong, P.; Israelow, B.; Lucas, C.; Klein, J.; Silva, J.; Mao, T.; Oh, J.E.; Tokuyama, M.; et al. Sex differences in immune responses that underlie COVID-19 disease outcomes. Nature 2020, 588, 315–320. [Google Scholar] [CrossRef] [PubMed]
  3. Kullar, R.; Marcelin, J.R.; Swartz, T.H.; Piggott, D.A.; Macias Gil, R.; Mathew, T.A.; Tan, T. Racial disparity of Coronavirus Disease 2019 in African American communities. J. Infect. Dis. 2020, 222, 890–893. [Google Scholar] [CrossRef]
  4. Navaratnam, A.V.; Gray, W.K.; Day, J.; Wendon, J.; Briggs, T.W.R. Patient factors and temporal trends associated with COVID-19 in-hospital mortality in England: An observational study using administrative data. Lancet Respir. Med. 2021, 9, 397–406. [Google Scholar] [CrossRef] [PubMed]
  5. Bennett, T.D.; Moffitt, R.A.; Hajagos, J.G.; Amor, B.; Anand, A.; Bissell, M.M.; Bradwell, K.R.; Bremer, C.; Byrd, J.B.; Denham, A.; et al. Clinical Characterization and Prediction of Clinical Severity of SARS-CoV-2 Infection Among US Adults Using Data from the US National COVID Cohort Collaborative. JAMA Netw. Open 2021, 4, e2116901. [Google Scholar] [CrossRef] [PubMed]
  6. Wolday, D.; Gebrecherkos, T.; Arefaine, Z.G.; Kiros, Y.K.; Gebreegzabher, A.; Tasew, G.; Abdulkader, M.; Abraha, H.E.; Desta, A.A.; Hailu, A.; et al. Effect of co-infection with intestinal parasites on COVID-19 severity: A prospective observational cohort study. eClinicalMedicine 2021, 39, 101054. [Google Scholar] [CrossRef]
  7. Lauring, A.S.; Tenforde, M.W.; Chappell, J.D.; Gaglani, M.; Ginde, A.A.; McNeal, T.; Ghamande, S.; Douin, D.J.; Talbot, H.K.; Casey, J.D.; et al. Clinical severity of, and effectiveness of mRNA vaccines against, COVID-19 from omicron, delta, and alpha SARS-CoV-2 variants in the United States: Prospective observational study. BMJ 2022, 376, e069761. [Google Scholar] [CrossRef]
  8. Zhang, Q.; Bastard, P.; Karbuz, A.; Gervais, A.; Tayoun, A.A.; Aiuti, A.; Belot, A.; Bolze, A.; Gaudet, A.; Bondarenko, A.; et al. Human genetic and immunological determinants of critical COVID-19 pneumonia. Nature 2022, 603, 587–598. [Google Scholar] [CrossRef]
  9. Dendrou, C.A.; Petersen, J.; Rossjohn, J.; Fugger, L. HLA variation and disease. Nat. Rev. Immunol. 2018, 18, 325–339. [Google Scholar] [CrossRef]
  10. Stephens, H.A.F.; Klaythong, R.; Sirikong, M.; Vaughn, D.W.; Green, S.; Kalayanarooj, S.; Endy, T.P.; Libraty, D.H.; Nisalak, A.; Innis, B.; et al. HLA-A and -B allele associations with secondary dengue virus infections correlate with disease severity and the infecting viral serotype in ethnic Thais. Tissue Antigens 2002, 60, 309–318. [Google Scholar] [CrossRef]
  11. Jiang, Y.-G.; Wang, Y.-M.; Liu, T.-H.; Liu, J. Association between HLA class II gene and susceptibility or resistance to chronic hepatitis B. World J. Gastroenterol. 2003, 9, 2221–2225. [Google Scholar] [CrossRef] [PubMed]
  12. Luo, M. Natural Immunity against HIV-1: Progression of Understanding after Association Studies. Viruses 2022, 14, 1243. [Google Scholar] [CrossRef] [PubMed]
  13. Harishankar, M.; Selvaraj, P.; Bethunaickan, R. Influence of Genetic Polymorphism Towards Pulmonary Tuberculosis Susceptibility. Front. Med. 2018, 5, 213–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kano, F.S.; Souza-Silva, F.A.; Torres, L.M.; Lima, B.A.S.; Sousa, T.N.; Alves, J.R.S.; Rocha, R.S.; Fontes, C.J.F.; Sanchez, B.A.M.; Adams, J.H.; et al. The Presence, Persistence and Functional Properties of Plasmodium vivax Duffy Binding Protein II Antibodies Are Influenced by HLA Class II Allelic Variants. PLoS Negl. Trop. Dis. 2016, 10, e0005177. [Google Scholar] [CrossRef] [Green Version]
  15. Ovsyannikova, I.G.; Pankratz, V.S.; Vierkant, R.A.; Jacobson, R.M.; Poland, G.A. Human Leukocyte Antigen Haplotypes in the Genetic Control of Immune Response to Measles-Mumps-Rubella Vaccine. J. Infect. Dis. 2006, 193, 655–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ovsyannikova, I.G.; Vierkant, R.A.; Pankratz, V.S.; Jacobson, R.M.; Poland, G.A. Human Leukocyte Antigen Genotypes in the Genetic Control of Adaptive Immune Responses to Smallpox Vaccine. J. Infect. Dis. 2011, 203, 1546–1555. [Google Scholar] [CrossRef]
  17. Liu, Y.; Guo, T.; Yu, Q.; Zhang, H.; Du, J.; Zhang, Y.; Xia, S.; Yang, H.; Li, Q. Association of human leukocyte antigen alleles and supertypes with immunogenicity of oral rotavirus vaccine given to infants in China. Medicine 2018, 97, e12706. [Google Scholar] [CrossRef]
  18. Png, E.; Thalamuthu, A.; Ong, R.T.; Snippe, H.; Boland, G.J.; Seielstad, M. A genome-wide association study of hepatitis B vaccine response in an Indonesian population reveals multiple independent risk variants in the HLA region. Hum. Mol. Genet. 2011, 20, 3893–3898. [Google Scholar] [CrossRef]
  19. Pan, L.; Zhang, L.; Zhang, W.; Wu, X.; Li, Y.; Yan, B.; Zhu, X.; Liu, X.; Yang, C.; Xu, J.; et al. A genome-wide association study identifies polymorphisms in the HLA-DR region associated with non-response to hepatitis B vaccination in Chinese Han populations. Hum. Mol. Genet. 2013, 23, 2210–2219. [Google Scholar] [CrossRef] [Green Version]
  20. Nishida, N.; Sugiyama, M.; Sawai, H.; Nishina, S.; Sakai, A.; Ohashi, J.; Khor, S.; Kakisaka, K.; Tsuchiura, T.; Hino, K.; et al. Key HLA-DRB1-DQB1 haplotypes and role of the BTNL2 gene for response to a hepatitis B vaccine. Hepatology 2018, 68, 848–858. [Google Scholar] [CrossRef] [Green Version]
  21. Chung, S.; Roh, E.Y.; Park, B.; Lee, Y.; Shin, S.; Yoon, J.H.; Song, E.Y. GWAS identifying HLA-DPB1 gene variants associated with responsiveness to hepatitis B virus vaccination in Koreans: Independent association of HLA-DPB1*04:02 possessing rs1042169 G-rs9277355 C-rs9277356 A. J. Viral Hepat. 2019, 26, 1318–1329. [Google Scholar] [CrossRef] [PubMed]
  22. Zhong, S.; Wei, H.; Li, M.; Cheng, Y.; Wen, S.; Wang, D.; Shu, Y. Single Nucleotide Polymorphisms in the Human Leukocyte Antigen Region Are Associated with Hemagglutination Inhibition Antibody Response to Influenza Vaccine. Front. Genet. 2022, 13, 790914. [Google Scholar] [CrossRef] [PubMed]
  23. Pairo-Castineira, E.; Clohisey, S.; Klaric, L.; Bretherick, A.D.; Rawlik, K.; Pasko, D.; Walker, S.; Parkinson, N.; Fourman, M.H.; Russell, C.D.; et al. Genetic mechanisms of critical illness in COVID-19. Nature 2021, 591, 92–98. [Google Scholar] [CrossRef] [PubMed]
  24. Niemi, M.E.K.; Daly, M.J.; Ganna, A. The human genetic epidemiology of COVID-19. Nat. Rev. Genet. 2022, 23, 533–546. [Google Scholar] [CrossRef] [PubMed]
  25. Zepeda-Cervantes, J.; Martínez-Flores, D.; Ramírez-Jarquín, J.O.; Tecalco-Cruz, C.; Alavez-Pérez, N.S.; Vaca, L.; Sarmiento-Silva, R.E. Implications of the Immune Polymorphisms of the Host and the Genetic Variability of SARS-CoV-2 in the Development of COVID-19. Viruses 2022, 14, 94. [Google Scholar] [CrossRef] [PubMed]
  26. Carsetti, R.; Zaffina, S.; Mortari, E.P.; Terreri, S.; Corrente, F.; Capponi, C.; Palomba, P.; Mirabella, M.; Cascioli, S.; Palange, P.; et al. Different Innate and Adaptive Immune Responses to SARS-CoV-2 Infection of Asymptomatic, Mild, and Severe Cases. Front. Immunol. 2020, 11, 610300. [Google Scholar] [CrossRef]
  27. Hall, V.; Foulkes, S.; Insalata, F.; Kirwan, P.; Saei, A.; Atti, A.; Wellington, E.; Khawam, J.; Munro, K.; Cole, M.; et al. Protection against SARS-CoV-2 after Covid-19 Vaccination and Previous Infection. N. Engl. J. Med. 2022, 386, 1207–1220. [Google Scholar] [CrossRef]
  28. Rose, R.; Neumann, F.; Grobe, O.; Lorentz, T.; Fickenscher, H.; Krumbholz, A. Humoral immune response after different SARS-CoV-2 vaccination regimens. BMC Med. 2022, 20, 31. [Google Scholar] [CrossRef]
  29. Barin, B.; Kasap, U.; Selçuk, F.; Volkan, E.; Uluçkan, O. Comparison of SARS-CoV-2 anti-spike receptor binding domain IgG antibody responses after CoronaVac, BNT162b2, ChAdOx1 COVID-19 vaccines, and a single booster dose: A prospective, longitudinal population-based study. Lancet Microbe 2022, 3, e274–e283. [Google Scholar] [CrossRef]
  30. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.-H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef]
  31. Tay, M.Z.; Poh, C.M.; Rénia, L.; Macary, P.A.; Ng, L.F.P. The trinity of COVID-19: Immunity, inflammation and intervention. Nat. Rev. Immunol. 2020, 20, 363–374. [Google Scholar] [CrossRef] [PubMed]
  32. Nelde, A.; Bilich, T.; Heitmann, J.S.; Maringer, Y.; Salih, H.R.; Roerden, M.; Lübke, M.; Bauer, J.; Rieth, J.; Wacker, M.; et al. SARS-CoV-2-derived peptides define heterologous and COVID-19-induced T cell recognition. Nat. Immunol. 2020, 22, 74–85. [Google Scholar] [CrossRef] [PubMed]
  33. Nagler, A.; Kalaora, S.; Barbolin, C.; Gangaev, A.; Ketelaars, S.L.; Alon, M.; Pai, J.; Benedek, G.; Yahalom-Ronen, Y.; Erez, N.; et al. Identification of presented SARS-CoV-2 HLA class I and HLA class II peptides using HLA peptidomics. Cell Rep. 2021, 35, 109305. [Google Scholar] [CrossRef]
  34. Grifoni, A.; Weiskopf, D.; Ramirez, S.I.; Mateus, J.; Dan, J.M.; Moderbacher, C.R.; Rawlings, S.A.; Sutherland, A.; Premkumar, L.; Jadi, R.S.; et al. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020, 181, 1489–1501.e1415. [Google Scholar] [CrossRef] [PubMed]
  35. Tarke, A.; Sidney, J.; Kidd, C.K.; Dan, J.M.; Ramirez, S.I.; Yu, E.D.; Mateus, J.; da Silva Antunes, R.; Moore, E.; Rubiro, P.; et al. Comprehensive analysis of T cell immunodominance and immunoprevalence of SARS-CoV-2 epitopes in COVID-19 cases. Cell Rep. Med. 2021, 2, 100204. [Google Scholar] [CrossRef] [PubMed]
  36. Tan, A.T.; Linster, M.; Tan, C.W.; Le Bert, N.; Ni Chia, W.; Kunasegaran, K.; Zhuang, Y.; Tham, C.Y.L.; Chia, A.; Smith, G.J.D.; et al. Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients. Cell Rep. 2021, 34, 108728. [Google Scholar] [CrossRef] [PubMed]
  37. Juno, J.A.; Tan, H.-X.; Lee, W.S.; Reynaldi, A.; Kelly, H.G.; Wragg, K.; Esterbauer, R.; Kent, H.E.; Batten, C.J.; Mordant, F.L.; et al. Humoral and circulating follicular helper T cell responses in recovered patients with COVID-19. Nat. Med. 2020, 26, 1428–1434. [Google Scholar] [CrossRef]
  38. Boppana, S.; Qin, K.; Files, J.K.; Russell, R.M.; Stoltz, R.; Bibollet-Ruche, F.; Bansal, A.; Erdmann, N.; Hahn, B.H.; Goepfert, P.A. SARS-CoV-2-specific circulating T follicular helper cells correlate with neutralizing antibodies and increase during early convalescence. PLoS Pathog. 2021, 17, e1009761. [Google Scholar] [CrossRef]
  39. Khoury, D.S.; Cromer, D.; Reynaldi, A.; Schlub, T.E.; Wheatley, A.K.; Juno, J.A.; Subbarao, K.; Kent, S.J.; Triccas, J.A.; Davenport, M.P. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat. Med. 2021, 27, 1205–1211. [Google Scholar] [CrossRef]
  40. Suthar, M.S.; Zimmerman, M.; Kauffman, R.; Mantus, G.; Linderman, S.; Vanderheiden, A.; Nyhoff, L.; Davis, C.; Adekunle, S.; Affer, M.; et al. Rapid generation of neutralizing antibody responses in COVID-19 patients. Cell Rep. Med. 2020, 1, 100040. [Google Scholar] [CrossRef]
  41. Altmann, D.M.; Boyton, R.J. SARS-CoV-2 T cell immunity: Specificity, function, durability, and role in protection. Sci. Immunol. 2020, 5, eabd6160. [Google Scholar] [CrossRef] [PubMed]
  42. Peng, Y.; Mentzer, A.J.; Liu, G.; Yao, X.; Yin, Z.; Dong, D.; Dejnirattisai, W.; Rostron, T.; Supasa, P.; Liu, C.; et al. Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat. Immunol. 2020, 21, 1336–1345. [Google Scholar] [CrossRef] [PubMed]
  43. Nguyen, D.C.; Lamothe, P.A.; Woodruff, M.C.; Saini, A.S.; Faliti, C.E.; Sanz, I.; Lee, F.E. COVID-19 and plasma cells: Is there long-lived protection? Immunol. Rev. 2022, 309, 40–63. [Google Scholar] [CrossRef] [PubMed]
  44. Cromer, D.; Juno, J.A.; Khoury, D.; Reynaldi, A.; Wheatley, A.K.; Kent, S.J.; Davenport, M.P. Prospects for durable immune control of SARS-CoV-2 and prevention of reinfection. Nat. Rev. Immunol. 2021, 21, 395–404. [Google Scholar] [CrossRef]
  45. Varga, Z.; Flammer, A.J.; Steiger, P.; Haberecker, M.; Andermatt, R.; Zinkernagel, A.S.; Mehra, M.R.; Schuepbach, R.A.; Ruschitzka, F.; Moch, H. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020, 395, 1417–1418. [Google Scholar] [CrossRef]
  46. Ackermann, M.; Verleden, S.E.; Kuehnel, M.; Haverich, A.; Welte, T.; Laenger, F.; Vanstapel, A.; Werlein, C.; Stark, H.; Tzankov, A.; et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N. Engl. J. Med. 2020, 383, 120–128. [Google Scholar] [CrossRef]
  47. Reusch, J.; Wagenhäuser, I.; Gabel, A.; Eggestein, A.; Höhn, A.; Lâm, T.; Frey, A.; Schubert-Unkmeir, A.; Dölken, L.; Frantz, S.; et al. Influencing factors of anti-SARS-CoV-2-spike-IgG antibody titers in healthcare workers: A cross-section study. J. Med. Virol. 2022, 95, e28300. [Google Scholar] [CrossRef]
  48. Karachaliou, M.; Moncunill, G.; Espinosa, A.; Castaño-Vinyals, G.; Rubio, R.; Vidal, M.; Jiménez, A.; Prados, E.; Carreras, A.; Cortés, B.; et al. SARS-CoV-2 infection, vaccination, and antibody response trajectories in adults: A cohort study in Catalonia. BMC Med. 2022, 20, 347. [Google Scholar] [CrossRef]
  49. Lindemann, M.; Klisanin, V.; Thümmler, L.; Fisenkci, N.; Tsachakis-Mück, N.; Ditschkowski, M.; Schwarzkopf, S.; Klump, H.; Reinhardt, H.C.; Horn, P.A.; et al. Humoral and Cellular Vaccination Responses against SARS-CoV-2 in Hematopoietic Stem Cell Transplant Recipients. Vaccines 2021, 9, 1075. [Google Scholar] [CrossRef]
  50. Dayam, R.M.; Law, J.C.; Goetgebuer, R.L.; Chao, G.Y.; Abe, K.T.; Sutton, M.; Finkelstein, N.; Stempak, J.M.; Pereira, D.; Croitoru, D.; et al. Accelerated waning of immunity to SARS-CoV-2 mRNA vaccines in patients with immune-mediated inflammatory diseases. J. Clin. Investig. 2022, 7, e159721. [Google Scholar] [CrossRef]
  51. Moncunill, G.; Aguilar, R.; Ribes, M.; Ortega, N.; Rubio, R.; Salmerón, G.; Molina, M.J.; Vidal, M.; Barrios, D.; Mitchell, R.A.; et al. Determinants of early antibody responses to COVID-19 mRNA vaccines in a cohort of exposed and naïve healthcare workers. eBioMedicine 2022, 75, 103805. [Google Scholar] [CrossRef] [PubMed]
  52. Röltgen, K.; Powell, A.E.; Wirz, O.F.; Stevens, B.A.; Hogan, C.A.; Najeeb, J.; Hunter, M.; Wang, H.; Sahoo, M.K.; Huang, C.; et al. Defining the features and duration of antibody responses to SARS-CoV-2 infection associated with disease severity and outcome. Sci. Immunol. 2020, 5, eabe0240. [Google Scholar] [CrossRef] [PubMed]
  53. Błaszczuk, A.; Michalski, A.; Sikora, D.; Malm, M.; Drop, B.; Polz-Dacewicz, M. Antibody Response after SARS-CoV-2 Infection with the Delta and Omicron Variant. Vaccines 2022, 10, 1728. [Google Scholar] [CrossRef] [PubMed]
  54. Medits, I.; Springer, D.N.; Graninger, M.; Camp, J.V.; Höltl, E.; Aberle, S.W.; Traugott, M.T.; Hoepler, W.; Deutsch, J.; Lammel, O.; et al. Different Neutralization Profiles After Primary SARS-CoV-2 Omicron BA.1 and BA.2 Infections. Front. Immunol. 2022, 13, 946318. [Google Scholar] [CrossRef] [PubMed]
  55. Uprichard, S.L.; O’Brien, A.; Evdokimova, M.; Rowe, C.L.; Joyce, C.; Hackbart, M.; Cruz-Pulido, Y.E.; Cohen, C.A.; Rock, M.L.; Dye, J.M.; et al. Antibody Response to SARS-CoV-2 Infection and Vaccination in COVID-19-naïve and Experienced Individuals. Viruses 2022, 14, 370. [Google Scholar] [CrossRef] [PubMed]
  56. Zheutlin, A.; Ott, M.; Sun, R.; Zemlianskaia, N.; Meyer, C.S.; Rubel, M.; Hayden, J.; Neri, B.; Kamath, T.; Khan, N.; et al. Durability of Protection Post–Primary COVID-19 Vaccination in the United States. Vaccines 2022, 10, 1458. [Google Scholar] [CrossRef]
  57. Bates, T.A.; McBride, S.K.; Leier, H.C.; Guzman, G.; Lyski, Z.L.; Schoen, D.; Winders, B.; Lee, J.-Y.; Lee, D.X.; Messer, W.B.; et al. Vaccination before or after SARS-CoV-2 infection leads to robust humoral response and antibodies that effectively neutralize variants. Sci. Immunol. 2022, 7, eabn8014. [Google Scholar] [CrossRef]
  58. Reynolds, C.J.; Pade, C.; Gibbons, J.M.; Butler, D.K.; Otter, A.D.; Menacho, K.; Fontana, M.; Smit, A.; Sackville-West, J.E.; Cutino-Moguel, T.; et al. Prior SARS-CoV-2 infection rescues B and T cell responses to variants after first vaccine dose. Science 2021, 372, 1418–1423. [Google Scholar] [CrossRef]
  59. Olmstead, A.D.; Nikiforuk, A.M.; Schwartz, S.; Márquez, A.C.; Valadbeigy, T.; Flores, E.; Saran, M.; Goldfarb, D.M.; Hayden, A.; Masud, S.; et al. Characterizing Longitudinal Antibody Responses in Recovered Individuals Following COVID-19 Infection and Single-Dose Vaccination: A Prospective Cohort Study. Viruses 2022, 14, 2416. [Google Scholar] [CrossRef]
  60. Khor, S.-S.; Omae, Y.; Takeuchi, J.S.; Fukunaga, A.; Yamamoto, S.; Tanaka, A.; Matsuda, K.; Kimura, M.; Maeda, K.; Ueda, G.; et al. An Association Study of HLA with the Kinetics of SARS-CoV-2 Spike Specific IgG Antibody Responses to BNT162b2 mRNA Vaccine. Vaccines 2022, 10, 563. [Google Scholar] [CrossRef]
  61. Mentzer, A.J.; O’Connor, D.; Bibi, S.; Chelysheva, I.; Clutterbuck, E.A.; Demissie, T.; Dinesh, T.; Edwards, N.J.; Felle, S.; Feng, S.; et al. Human leukocyte antigen alleles associate with COVID-19 vaccine immunogenicity and risk of breakthrough infection. Nat. Med. 2022, 29, 147–157. [Google Scholar] [CrossRef] [PubMed]
  62. Astbury, S.; Reynolds, C.J.; Butler, D.K.; Muñoz-Sandoval, D.C.; Lin, K.; Pieper, F.P.; Otter, A.; Kouraki, A.; Cusin, L.; Nightingale, J.; et al. HLA-DR polymorphism in SARS-CoV-2 infection and susceptibility to symptomatic COVID-19. Immunology 2022, 166, 68–77. [Google Scholar] [CrossRef] [PubMed]
  63. Gutiérrez-Bautista, J.F.; Sampedro, A.; Gómez-Vicente, E.; Rodríguez-Granger, J.; Reguera, J.A.; Cobo, F.; Ruiz-Cabello, F.; López-Nevot, M. HLA Class II Polymorphism and Humoral Immunity Induced by the SARS-CoV-2 mRNA-1273 Vaccine. Vaccines 2022, 10, 402. [Google Scholar] [CrossRef] [PubMed]
  64. Higuchi, T.; Oka, S.; Furukawa, H.; Tohma, S. Associations of HLA Polymorphisms with Anti-SARS-CoV-2 Spike and Neutralizing Antibody Titers in Japanese Rheumatoid Arthritis Patients Vaccinated with BNT162b2. Vaccines 2023, 11, 404. [Google Scholar] [CrossRef]
  65. Crocchiolo, R.; Gallina, A.M.; Pani, A.; Campisi, D.; Cento, V.; Sacchi, N.; Miotti, V.; Gagliardi, O.M.; D’Amico, F.; Vismara, C.; et al. Polymorphism of the HLA system and weak antibody response to BNT162b2 mRNA vaccine. HLA 2022, 99, 183–191. [Google Scholar] [CrossRef]
  66. Fischer, J.C.; Schmidt, A.G.; Bölke, E.; Uhrberg, M.; Keitel, V.; Feldt, T.; Jensen, B.; Häussinger, D.; Adams, O.; Schneider, E.M.; et al. Association of HLA genotypes, AB0 blood type and chemokine receptor 5 mutant CD195 with the clinical course of COVID-19. Eur. J. Med. Res. 2021, 26, 107. [Google Scholar] [CrossRef]
  67. Weidner, L.; Kalser, J.; Kreil, T.R.; Jungbauer, C.; Mayr, W.R. Neutralizing Antibodies against SARS-CoV-2 and HLA Class I and II Polymorphism. Transfus. Med. Hemother. 2021, 48, 173–174. [Google Scholar] [CrossRef]
  68. Ragone, C.; Meola, S.; Fiorillo, P.C.; Penta, R.; Auriemma, L.; Tornesello, M.L.; Miscio, L.; Cavalcanti, E.; Botti, G.; Buonaguro, F.M.; et al. HLA Does Not Impact on Short-Medium-Term Antibody Response to Preventive Anti-SARS-Cov-2 Vaccine. Front. Immunol. 2021, 12, 734689. [Google Scholar] [CrossRef]
  69. Gerhards, C.; Kittel, M.; Ast, V.; Bugert, P.; Froelich, M.F.; Hetjens, M.; Haselmann, V.; Neumaier, M.; Thiaucourt, M. Humoral SARS-CoV-2 Immune Response in COVID-19 Recovered Vaccinated and Unvaccinated Individuals Related to Post-COVID-Syndrome. Viruses 2023, 15, 454. [Google Scholar] [CrossRef]
  70. Kiyotani, K.; Toyoshima, Y.; Nemoto, K.; Nakamura, Y. Bioinformatic prediction of potential T cell epitopes for SARS-Cov-2. J. Hum. Genet. 2020, 65, 569–575. [Google Scholar] [CrossRef]
  71. Romero-López, J.P.; Carnalla-Cortés, M.; Pacheco-Olvera, D.L.; Ocampo-Godínez, J.M.; Oliva-Ramírez, J.; Moreno-Manjón, J.; Bernal-Alferes, B.; López-Olmedo, N.; García-Latorre, E.; Domínguez-López, M.L.; et al. A bioinformatic prediction of antigen presentation from SARS-CoV-2 spike protein revealed a theoretical correlation of HLA-DRB1*01 with COVID-19 fatality in Mexican population: An ecological approach. J. Med. Virol. 2020, 93, 2029–2038. [Google Scholar] [CrossRef] [PubMed]
  72. Toyoshima, Y.; Nemoto, K.; Matsumoto, S.; Nakamura, Y.; Kiyotani, K. SARS-CoV-2 genomic variations associated with mortality rate of COVID-19. J. Hum. Genet. 2020, 65, 1075–1082. [Google Scholar] [CrossRef]
  73. Shkurnikov, M.; Nersisyan, S.; Jankevic, T.; Galatenko, A.; Gordeev, I.; Vechorko, V.; Tonevitsky, A. Association of HLA Class I Genotypes with Severity of Coronavirus Disease-19. Front. Immunol. 2021, 12, 641900. [Google Scholar] [CrossRef]
  74. Tomita, Y.; Ikeda, T.; Sato, R.; Sakagami, T. Association between HLA gene polymorphisms and mortality of COVID-19: An in silico analysis. Immun. Inflamm. Dis. 2020, 8, 684–694. [Google Scholar] [CrossRef] [PubMed]
  75. Pretti, M.A.M.; Galvani, R.G.; Vieira, G.F.; Bonomo, A.; Bonamino, M.H.; Boroni, M. Class I HLA Allele Predicted Restricted Antigenic Coverages for Spike and Nucleocapsid Proteins Are Associated with Deaths Related to COVID-19. Front. Immunol. 2020, 11, 565730. [Google Scholar] [CrossRef] [PubMed]
  76. Ishii, T. Human Leukocyte Antigen (HLA) Class I Susceptible Alleles Against COVID-19 Increase Both Infection and Severity Rate. Cureus 2020, 12, e12239. [Google Scholar] [CrossRef]
  77. Naemi, F.M.A.; Al-Adwani, S.; Al-Khatabi, H.; Al-Nazawi, A. Association between the HLA genotype and the severity of COVID-19 infection among South Asians. J. Med. Virol. 2021, 93, 4430–4437. [Google Scholar] [CrossRef] [PubMed]
  78. Littera, R.; Campagna, M.; Deidda, S.; Angioni, G.; Cipri, S.; Melis, M.; Firinu, D.; Santus, S.; Lai, A.; Porcella, R.; et al. Human Leukocyte Antigen Complex and Other Immunogenetic and Clinical Factors Influence Susceptibility or Protection to SARS-CoV-2 Infection and Severity of the Disease Course. The Sardinian Experience. Front. Immunol. 2020, 11, 605688. [Google Scholar] [CrossRef]
  79. Yarmarkovich, M.; Warrington, J.M.; Farrel, A.; Maris, J.M. Identification of SARS-CoV-2 Vaccine Epitopes Predicted to Induce Long-Term Population-Scale Immunity. Cell Rep. Med. 2020, 1, 100036. [Google Scholar] [CrossRef]
  80. Lorente, L.; Martín, M.; Franco, A.; Barrios, Y.; Cáceres, J.; Solé-Violán, J.; Perez, A.; Ramos, J.M.Y.; Ramos-Gómez, L.; Ojeda, N.; et al. HLA genetic polymorphisms and prognosis of patients with COVID-19. Med. Intensiv. 2021, 45, 96–103. [Google Scholar] [CrossRef]
  81. Wang, F.; Huang, S.; Gao, R.; Zhou, Y.; Lai, C.; Li, Z.; Xian, W.; Qian, X.; Li, Z.; Huang, Y.; et al. Initial whole-genome sequencing and analysis of the host genetic contribution to COVID-19 severity and susceptibility. Cell Discov. 2020, 6, 83. [Google Scholar] [CrossRef] [PubMed]
  82. Khor, S.-S.; Omae, Y.; Nishida, N.; Sugiyama, M.; Kinoshita, N.; Suzuki, T.; Suzuki, M.; Suzuki, S.; Izumi, S.; Hojo, M.; et al. HLA-A*11:01:01:01, HLA-C*12:02:02:01-HLA-B*52:01:02:02, Age and Sex Are Associated with Severity of Japanese COVID-19 with Respiratory Failure. Front. Immunol. 2021, 12, 658570. [Google Scholar] [CrossRef] [PubMed]
  83. Warren, R.L.; Birol, I. Retrospective in silico HLA predictions from COVID-19 patients reveal alleles associated with disease prognosis. MedRxiv 2020. [Google Scholar] [CrossRef]
  84. Sakuraba, A.; Haider, H.; Sato, T. Population Difference in Allele Frequency of HLA-C*05 and Its Correlation with COVID-19 Mortality. Viruses 2020, 12, 1333. [Google Scholar] [CrossRef]
  85. Pisanti, S.; Deelen, J.; Gallina, A.M.; Caputo, M.; Citro, M.; Abate, M.; Sacchi, N.; Vecchione, C.; Martinelli, R. Correlation of the two most frequent HLA haplotypes in the Italian population to the differential regional incidence of COVID-19. J. Transl. Med. 2020, 18, 352. [Google Scholar] [CrossRef] [PubMed]
  86. Warren, R.L.; Birol, I. HLA predictions from the bronchoalveolar lavage fluid and blood samples of eight COVID-19 patients at the pandemic onset. Bioinformatics 2020, 36, 5271–5273. [Google Scholar] [CrossRef] [PubMed]
  87. Nguyen, A.; David, J.K.; Maden, S.K.; Wood, M.A.; Weeder, B.R.; Nellore, A.; Thompson, R.F. Human Leukocyte Antigen Susceptibility Map for Severe Acute Respiratory Syndrome Coronavirus 2. J. Virol. 2020, 94, e00510-20. [Google Scholar] [CrossRef] [Green Version]
  88. Correale, P.; Mutti, L.; Pentimalli, F.; Baglio, G.; Saladino, R.; Sileri, P.; Giordano, A. HLA-B*44 and C*01 Prevalence Correlates with Covid19 Spreading across Italy. Int. J. Mol. Sci. 2020, 21, 5205. [Google Scholar] [CrossRef]
  89. Novelli, A.; Andreani, M.; Biancolella, M.; Liberatoscioli, L.; Passarelli, C.; Colona, V.L.; Rogliani, P.; Leonardis, F.; Campana, A.; Carsetti, R.; et al. HLA allele frequencies and susceptibility to COVID-19 in a group of 99 Italian patients. HLA 2020, 96, 610–614. [Google Scholar] [CrossRef]
  90. Poulton, K.; Wright, P.; Hughes, P.; Savic, S.; Smith, M.W.; Guiver, M.; Morton, M.; Van Dellen, D.; Tholouli, E.; Wynn, R.; et al. A role for human leucocyte antigens in the susceptibility to SARS-Cov-2 infection observed in transplant patients. Int. J. Immunogenet. 2020, 47, 324–328. [Google Scholar] [CrossRef]
  91. Langton, D.J.; Bourke, S.C.; Lie, B.A.; Reiff, G.; Natu, S.; Darlay, R.; Burn, J.; Echevarria, C. The influence of HLA genotype on the severity of COVID-19 infection. HLA 2021, 98, 14–22. [Google Scholar] [CrossRef] [PubMed]
  92. Anzurez, A.; Naka, I.; Miki, S.; Nakayama-Hosoya, K.; Isshiki, M.; Watanabe, Y.; Nakamura-Hoshi, M.; Seki, S.; Matsumura, T.; Takano, T.; et al. Association of HLA-DRB1 *09:01 with severe COVID-19. HLA 2021, 98, 37–42. [Google Scholar] [CrossRef]
  93. Amoroso, A.; Magistroni, P.; Vespasiano, F.; Bella, A.; Bellino, S.; Puoti, F.; Alizzi, S.; Vaisitti, T.; Boros, S.; Grossi, P.A.; et al. HLA and AB0 Polymorphisms May Influence SARS-CoV-2 Infection and COVID-19 Severity. Transplantation 2021, 105, 193–200. [Google Scholar] [CrossRef]
  94. Savage, P.A.; Klawon, D.E.; Miller, C.H. Regulatory T Cell Development. Annu. Rev. Immunol. 2020, 38, 421–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Bauer, D.C.; Zadoorian, A.; Wilson, L.O.W.; Thorne, N.P. Melbourne Genomics Health Alliance Evaluation of computational programs to predict HLA genotypes from genomic sequencing data. Brief. Bioinform. 2016, 19, 179–187. [Google Scholar] [CrossRef] [Green Version]
  96. Liu, P.; Yao, M.; Gong, Y.; Song, Y.; Chen, Y.; Ye, Y.; Liu, X.; Li, F.; Dong, H.; Meng, R.; et al. Benchmarking the Human Leukocyte Antigen Typing Performance of Three Assays and Seven Next-Generation Sequencing-Based Algorithms. Front. Immunol. 2021, 12, 652258. [Google Scholar] [CrossRef] [PubMed]
  97. Liu, C.; Duffy, B.F.; Weimer, E.T.; Montgomery, M.C.; Jennemann, J.-E.; Hill, R.; Phelan, D.; Lay, L.; Parikh, B.A. Performance of a multiplexed amplicon-based next-generation sequencing assay for HLA typing. PLoS ONE 2020, 15, e0232050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Cereb, N.; Kim, H.R.; Ryu, J.; Yang, S.Y. Advances in DNA sequencing technologies for high resolution HLA typing. Hum. Immunol. 2015, 76, 923–927. [Google Scholar] [CrossRef] [PubMed]
  99. Duke, J.L.; Lind, C.; Mackiewicz, K.; Ferriola, D.; Papazoglou, A.; Gasiewski, A.; Heron, S.; Huynh, A.; McLaughlin, L.; Rogers, M.; et al. Determining performance characteristics of an NGS-based HLA typing method for clinical applications. HLA 2016, 87, 141–152. [Google Scholar] [CrossRef]
  100. Balz, V.; Fischer, J.; Krause, S.; Enczmann, J. A novel HLA-B variant, HLA-B*14:02:16, discovered by amplicon-based next-generation sequencing. HLA 2018, 92, 323–324. [Google Scholar] [CrossRef]
  101. Balz, V.; Krause, S.; Fischer, J.; Enczmann, J. More than 150 novel variants of HLA class I genes detected in German Stem Cell Donor Registry and UCLA International Cell Exchange samples. HLA 2018, 91, 187–194. [Google Scholar] [CrossRef] [PubMed]
  102. Cargou, M.; Ralazamahaleo, M.; Blouin, L.; Top, I.; Elsermans, V.; Andreani, M.; Guidicelli, G.; Visentin, J. Evaluation of the AllType kit for HLA typing using the Ion Torrent S5 XL platform. HLA 2019, 95, 30–39. [Google Scholar] [CrossRef]
  103. Gutiérrez-Bautista, J.F.; Rodriguez-Nicolas, A.; Rosales-Castillo, A.; López-Ruz, M.; Martín-Casares, A.M.; Fernández-Rubiales, A.; Anderson, P.; Garrido, F.; Ruiz-Cabello, F.; López-Nevot, M. Study of HLA-A, -B, -C, -DRB1 and -DQB1 polymorphisms in COVID-19 patients. J. Microbiol. Immunol. Infect. 2021, 55, 421–427. [Google Scholar] [CrossRef]
  104. McCarthy, S.; Das, S.; Kretzschmar, W.; Delaneau, O.; Wood, A.R.; Teumer, A.; Kang, H.M.; Fuchsberger, C.; Danecek, P.; Sharp, K.; et al. A reference panel of 64,976 haplotypes for genotype imputation. Nat. Genet. 2016, 48, 1279–1283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Luo, Y.; Kanai, M.; Choi, W.; Li, X.; Sakaue, S.; Yamamoto, K.; Ogawa, K.; Gutierrez-Arcelus, M.; Gregersen, P.K.; Stuart, P.E.; et al. A high-resolution HLA reference panel capturing global population diversity enables multi-ancestry fine-mapping in HIV host response. Nat. Genet. 2021, 53, 1504–1516. [Google Scholar] [CrossRef] [PubMed]
  106. Zheng, X.; Shen, J.; Cox, C.J.; Wakefield, J.C.; Ehm, M.G.; Nelson, M.R.; Weir, B.S. HIBAG—HLA genotype imputation with attribute bagging. Pharm. J. 2013, 14, 192–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Chitnis, N.; Clark, P.M.; Kamoun, M.; Stolle, C.; Johnson, F.B.; Monos, D.S. An Expanded Role for HLA Genes: HLA-B Encodes a microRNA that Regulates IgA and Other Immune Response Transcripts. Front. Immunol. 2017, 8, 583–594. [Google Scholar] [CrossRef]
  108. Colucci, M.; De Santis, E.; Totti, B.; Miroballo, M.; Tamiro, F.; Rossi, G.; Piepoli, A.; De Vincentis, G.; Greco, A.; Mangia, A.; et al. Associations between Allelic Variants of the Human IgH 3′ Regulatory Region 1 and the Immune Response to BNT162b2 mRNA Vaccine. Vaccines 2021, 9, 1207. [Google Scholar] [CrossRef]
  109. Čiučiulkaitė, I.; Möhlendick, B.; Thümmler, L.; Fisenkci, N.; Elsner, C.; Dittmer, U.; Siffert, W.; Lindemann, M. GNB3 c.825c>T polymorphism influences T-cell but not antibody response following vaccination with the mRNA-1273 vaccine. Front. Genet. 2022, 13, 932043. [Google Scholar] [CrossRef]
  110. Gemmati, D.; Longo, G.; Gallo, I.; Silva, J.A.; Secchiero, P.; Zauli, G.; Hanau, S.; Passaro, A.; Pellegatti, P.; Pizzicotti, S.; et al. Host genetics impact on SARS-CoV-2 vaccine-induced immunoglobulin levels and dynamics: The role of TP53, ABO, APOE, ACE2, HLA-A, and CRP genes. Front. Genet. 2022, 13, 1028081. [Google Scholar] [CrossRef]
  111. Kousathanas, A.; Pairo-Castineira, E.; Rawlik, K.; Stuckey, A.; Odhams, C.A.; Walker, S.; Russell, C.D.; Malinauskas, T.; Wu, Y.; Millar, J.; et al. Whole-genome sequencing reveals host factors underlying critical COVID-19. Nature 2022, 607, 97–103. [Google Scholar] [CrossRef] [PubMed]
  112. Fallerini, C.; Daga, S.; Mantovani, S.; Benetti, E.; Picchiotti, N.; Francisci, D.; Paciosi, F.; Schiaroli, E.; Baldassarri, M.; Fava, F.; et al. Association of Toll-like receptor 7 variants with life-threatening COVID-19 disease in males: Findings from a nested case-control study. eLife 2021, 10, e67569. [Google Scholar] [CrossRef] [PubMed]
  113. Butler-Laporte, G.; Povysil, G.; Kosmicki, J.A.; Cirulli, E.T.; Drivas, T.; Furini, S.; Saad, C.; Schmidt, A.; Olszewski, P.; Korotko, U.; et al. Exome-wide association study to identify rare variants influencing COVID-19 outcomes: Results from the Host Genetics Initiative. PLoS Genet. 2022, 18, e1010367. [Google Scholar] [CrossRef]
  114. Arora, P.; Sidarovich, A.; Krüger, N.; Kempf, A.; Nehlmeier, I.; Graichen, L.; Moldenhauer, A.-S.; Winkler, M.S.; Schulz, S.; Jäck, H.-M.; et al. B.1.617.2 enters and fuses lung cells with increased efficiency and evades antibodies induced by infection and vaccination. Cell Rep. 2021, 37, 109825. [Google Scholar] [CrossRef]
  115. Madhi, S.A.; Baillie, V.; Cutland, C.L.; Voysey, M.; Koen, A.L.; Fairlie, L.; Padayachee, S.D.; Dheda, K.; Barnabas, S.L.; Bhorat, Q.E.; et al. Efficacy of the ChAdOx1 nCoV-19 Covid-19 Vaccine against the B.1.351 Variant. N. Engl. J. Med. 2021, 384, 1885–1898. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, Z.; Schmidt, F.; Weisblum, Y.; Muecksch, F.; Barnes, C.O.; Finkin, S.; Schaefer-Babajew, D.; Cipolla, M.; Gaebler, C.; Lieberman, J.A.; et al. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. Nature 2021, 592, 616–622. [Google Scholar] [CrossRef]
  117. Wibmer, C.K.; Ayres, F.; Hermanus, T.; Madzivhandila, M.; Kgagudi, P.; Oosthuysen, B.; Lambson, B.E.; De Oliveira, T.; Vermeulen, M.; Van der Berg, K.; et al. SARS-CoV-2 501Y.V2 Escapes Neutralization by South African COVID-19 Donor Plasma. Nat. Med. 2021, 27, 622–625. [Google Scholar] [CrossRef]
  118. Emmelot, M.E.; Vos, M.; Boer, M.C.; Rots, N.Y.; van Els, C.A.C.M.; Kaaijk, P. SARS-CoV-2 Omicron BA.4/BA.5 Mutations in Spike Leading to T Cell Escape in Recently Vaccinated Individuals. Viruses 2022, 15, 101. [Google Scholar] [CrossRef]
  119. Xiao, C.; Mao, L.; Wang, Z.; Gao, L.; Zhu, G.; Su, J.; Chen, X.; Yuan, J.; Hu, Y.; Yin, Z.; et al. SARS-CoV-2 variant B.1.1.7 caused HLA-A2+ CD8+ T cell epitope mutations for impaired cellular immune response. iScience 2022, 25, 103934. [Google Scholar] [CrossRef]
  120. Nersisyan, S.; Zhiyanov, A.; Shkurnikov, M.; Tonevitsky, A. T-CoV: A comprehensive portal of HLA-peptide interactions affected by SARS-CoV-2 mutations. Nucleic Acids Res. 2021, 50, D883–D887. [Google Scholar] [CrossRef]
  121. Nersisyan, S.; Zhiyanov, A.; Zakharova, M.; Ishina, I.; Kurbatskaia, I.; Mamedov, A.; Galatenko, A.; Shkurnikov, M.; Gabibov, A.; Tonevitsky, A. Alterations in SARS-CoV-2 Omicron and Delta peptides presentation by HLA molecules. PeerJ 2022, 10, e13354. [Google Scholar] [CrossRef] [PubMed]
  122. Chen, L.-C.; Nersisyan, S.; Wu, C.-J.; Chang, C.-M.; Tonevitsky, A.; Guo, C.-L.; Chang, W.-C. On the peptide binding affinity changes in population-specific HLA repertoires to the SARS-CoV-2 variants Delta and Omicron. J. Autoimmun. 2022, 133, 102952. [Google Scholar] [CrossRef] [PubMed]
  123. Taher, J.; Mighton, C.; Chowdhary, S.; Casalino, S.; Frangione, E.; Arnoldo, S.; Bearss, E.; Binnie, A.; Bombard, Y.; Borgundvaag, B.; et al. Implementation of serological and molecular tools to inform COVID-19 patient management: Protocol for the GENCOV prospective cohort study. BMJ Open 2021, 11, e052842. [Google Scholar] [CrossRef] [PubMed]
Figure 1. HLA variation determines SARS-CoV-2-specific antibody response. In this example, we present different scenarios suggesting differential SARS-CoV-2-specific antibody response impacted by variation in the HLA class II molecule. (A) MHC-II molecule binding to SARS-CoV-2 peptides induces efficient CD4+ T cell and antibody response leading to viral neutralization, ADCC, and the death of SARS-CoV-2 infected cells. (B) Polymorphic MHC-II molecule binding to viral peptide fails to stimulate CD4+ T cells and results in low antibody titer, lack of ADCC, and ADP. Abbreviations: Ab: antibody; ADCC: antibody-dependent cellular (NK cell) cytotoxicity; ADP: antibody-dependent phagocytosis; HLA: human leukocyte antigen; MØ: macrophage; MHC: major histocompatibility complex; nAbs: neutralizing antibodies; TH0: CD4+ helper cells; TH: T helper; TFH: T follicular helper cells. Figure made using BioRender.
Figure 1. HLA variation determines SARS-CoV-2-specific antibody response. In this example, we present different scenarios suggesting differential SARS-CoV-2-specific antibody response impacted by variation in the HLA class II molecule. (A) MHC-II molecule binding to SARS-CoV-2 peptides induces efficient CD4+ T cell and antibody response leading to viral neutralization, ADCC, and the death of SARS-CoV-2 infected cells. (B) Polymorphic MHC-II molecule binding to viral peptide fails to stimulate CD4+ T cells and results in low antibody titer, lack of ADCC, and ADP. Abbreviations: Ab: antibody; ADCC: antibody-dependent cellular (NK cell) cytotoxicity; ADP: antibody-dependent phagocytosis; HLA: human leukocyte antigen; MØ: macrophage; MHC: major histocompatibility complex; nAbs: neutralizing antibodies; TH0: CD4+ helper cells; TH: T helper; TFH: T follicular helper cells. Figure made using BioRender.
Viruses 15 00906 g001
Figure 2. Heat-map showing the association between HLA variants and antibody responses or epitope binding affinity. Associations are depicted arbitrarily as high (scaled 1, yellow boxes), low (scaled 0.5, red boxes), or no association (scaled 0, dark-blue boxes). Gray boxes indicate undetermined values. References shown are for antibody response* or epitope binding affinity** studies.
Figure 2. Heat-map showing the association between HLA variants and antibody responses or epitope binding affinity. Associations are depicted arbitrarily as high (scaled 1, yellow boxes), low (scaled 0.5, red boxes), or no association (scaled 0, dark-blue boxes). Gray boxes indicate undetermined values. References shown are for antibody response* or epitope binding affinity** studies.
Viruses 15 00906 g002
Table 1. Studies on HLA variants and association with antibody responses.
Table 1. Studies on HLA variants and association with antibody responses.
Study Cohort CharacteristicsAntibody ResponseReferences
Infection StatusVaccinationNumber of
Individuals
Date Antibody
Response
Assessed
Antibody
Response
Assessed
No prior
infection
BNT162b21007 and 39 days after
second vaccine dose
Anti-RBD IgG[60]
With or without prior
infection
ChAdOx1
or
ChAdOx1+ BNT162b2 or mRNA-1273 or NVX-CoV2373
1076
or
1677
28 days after first
vaccine dose
or
28 to 184 days after second vaccine dose
Anti-RBD and/or anti-S IgG[61]
With or without prior
infection
BNT162b2420UnknownAnti-RBD IgG[62]
No prior
infection
mRNA12738730 days after second vaccine doseAnti-S IgG[63]
No prior
infection
BNT162b287144 days after second vaccine doseAnti-RBD IgG and
Neutralizing
Antibodies
[64]
No prior
infection
BNT162b211114+ days after second vaccine doseAnti-S IgG[65]
Prior
infection
None119>46 to >97 days after end of symptomsAnti-RBD IgG, Anti-RBD IgA,
Anti-NP total Ig, and
Neutralizing
antibodies
[66]
Prior
Infection
None8426 and 61 days after
symptom onset
Neutralizing
antibodies
[67]
No prior
infection
BNT162b2562 weeks to 4 months after second vaccine doseAnti-RBD IgG[68]
Prior
infection
With or without BNT162b2 or ChAdOx14912 months after
PCR positivity
Anti-RBD IgG[69]
Abbreviations: Ig: immunoglobulin; NP: nucleocapsid protein; RBD: receptor-binding domain; S: spike antigen.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wolday, D.; Fung, C.Y.J.; Morgan, G.; Casalino, S.; Frangione, E.; Taher, J.; Lerner-Ellis, J.P. HLA Variation and SARS-CoV-2 Specific Antibody Response. Viruses 2023, 15, 906. https://doi.org/10.3390/v15040906

AMA Style

Wolday D, Fung CYJ, Morgan G, Casalino S, Frangione E, Taher J, Lerner-Ellis JP. HLA Variation and SARS-CoV-2 Specific Antibody Response. Viruses. 2023; 15(4):906. https://doi.org/10.3390/v15040906

Chicago/Turabian Style

Wolday, Dawit, Chun Yiu Jordan Fung, Gregory Morgan, Selina Casalino, Erika Frangione, Jennifer Taher, and Jordan P. Lerner-Ellis. 2023. "HLA Variation and SARS-CoV-2 Specific Antibody Response" Viruses 15, no. 4: 906. https://doi.org/10.3390/v15040906

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop