key: cord-0992836-35mfg8b7 authors: Alefishat, Eman; Jelinek, Herbert F.; Mousa, Mira; Tay, Guan K.; Alsafar, Habiba S title: Immune response to SARS-CoV-2 Variants: A focus on severity, susceptibility, and preexisting immunity date: 2022-01-13 journal: J Infect Public Health DOI: 10.1016/j.jiph.2022.01.007 sha: 4f33b4456947c77f97e4cc811e32807797f91f88 doc_id: 992836 cord_uid: 35mfg8b7 The heterogeneous phenotypes among patients with coronavirus disease 2019 (COVID-19) has drawn worldwide attention, especially those with severe symptoms without comorbid conditions. Immune responses to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative virus of COVID-19, occur mainly by the innate immune response via the interferon (IFN)-mediated pathways, and the adaptive immunity via the T lymphocyte and the antibody mediated pathways. The ability of the original Wuhan SARS-CoV-2 strain, and possibly more so with new emerging variants, to antagonize IFN-mediated antiviral responses can be behind the higher early viral load, higher transmissibility, and milder symptoms compared to SARS-CoV and are part of the continued clinical evolution of COVID-19. Since it first emerged, several variants of SARS-CoV-2 have been circulating worldwide. Variants that have the potential to elude natural or vaccine-mediated immunity are variants of concern. This review focuses on the main host factors that may explain the immune responses to SARS-CoV-2 and its variants in the context of susceptibility, severity, and preexisting immunity. Innate immunity is the first line response to protect host cells from viral infection. The features of this response elicit nonspecific immune reactions that determine the outcome of the infection and the heterogeneous clinical phenotypes [17] . Innate defense mechanisms were found to be sufficient to control SARS-CoV infection, in the absence of CD4 + T cells, CD8 + T cells and antibodies [18] [19] [20] . In fact, the response to SARS-CoV has been reported to be mainly through the innate inflammatory response, rather than the specific adaptive immune response, due to possibly a lack of cytokine activity [21] . However, viruses have evolved to either evade or inactivate the innate immune responses or lead to a hyperactivated inflammatory response [22] . SARS-CoVs are single-stranded RNA (ssRNA) viruses that stimulates innate immune responses and are excessive when compared to other viruses, such as influenza [23, 24] . Regions of the SARS-CoVs genome were shown to work as immunostimulants for several components of innate immunity through the activation of Toll-like receptors, such as TNF-α, IL-6 and IL-12 [25, 26] . In comparison to other ssRNA viruses, this immunostimulant activity is two-fold higher [23] . A major factor related to disease outcome is the ability to resolve the initial inflammatory response [27] , demonstrating that a strong innate immune response is not necessarily the best response [28] . Several components of the innate immune response have been found to play an important role in the progression and clinical presentation of COVID-19 infection. The innate immune response to SARS-CoV-2 infection is mediated via alveolar macrophages and dendritic cells which express pattern recognition receptors (PRR) which recognize Pathogen-Associated Molecular Patterns (PAMPs) and Damage-Associated Molecular Patterns (DAMPs) [29] . During this recognition, the NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome is activated [30, 31] . This activation of the NLRP3 inflammasome plays a vital role in the early stages of infection to efficiently limit virus replication by subsequent inflammation [32, 33] . Limited or dysfunctional NLRP3 inflammasome activation has an essential role in the pathogenesis of severe organ injury [31, [33] [34] [35] . Although activation of the NLRP3 inflammasome and its mediated inflammation is necessary for the defense against viruses, impaired/excessive activation can also mediate damage and adverse disease outcomes [31, [33] [34] [35] . In SARS-CoV-2 infection, despite the lack of data regarding any direct association between the NLRP3 inflammasome and COVID- 19 , several studies suggested the involvement of NLRP3 and cytokine storm and cell apoptosis [30, [36] [37] [38] [39] . The activation of inflammasome in macrophages, epithelial cells, and endothelial cells results in the releases of proinflammatory cytokines, most notably, IL-1β and IL-18, leading to the characteristic feature of neutrophilia and leukopenia seen in severe COVID-19 cases [40, 41] . Mannose-binding lectin (MBL) protein is also a contributor to pattern-recognition molecules and plays a critical role in the first-line host defense against SARS-CoV, before antibody production. MBL deficiency has been linked to an increased susceptibility to SARS-CoV infection [42] [43] [44] , and correlated with an increased genetic predisposition to SARS-CoV [43] . In COVID-19 patients, MBL pathway-mediated complement activation has been shown to contribute to thrombosis and coagulopathy in patients with severe COVID-19 [45] . In addition, Toll-Like Receptor (TLR) sense viral RNA that results in the activation of TLR3, TLR7, TLR8, and TLR9, and consequently activates the Nuclear Factor kappa B (NF-κB) pathway. A large number of pro-inflammatory cytokines play a major role in triggering virusinduced inflammation [46] . The increased secretion of the pro-inflammatory cytokines and chemokines recruits mainly monocytes and T lymphocytes, but not neutrophils to the infected site, elucidate the presentation of lymphopenia and the high neutrophil-lymphocyte ratio seen in most COVID-19 patients [47] . J o u r n a l P r e -p r o o f In most COVID-19 patients, recruited immune cells controls the infection in the pulmonary tissue that terminates the immune response, and leads to recovery from the viral infection. However, in a subset of patients, evasion of innate immunity occurs and a later excessive/dysfunctional immune response is triggered, culminating into a cytokine storm during which an unusual lack of IFN and a massive elevation of Granulocyte Colony-Stimulating Factor (G-CSF), IP-10, MCP-1, Macrophage Inflammatory Protein 1α (MIP-1α) and Tumor Necrosis Factor-alpha (TNF-α), IL-2, IL-6, IL-7, IL-10 are observed [47, 48] . The exact mechanism of how SARS-CoV-2 evades the innate immunity and causes excessive inflammatory response that result in a higher degree of viral load is yet to be determined, but antagonism of the interferon signaling pathway have been highlighted [47] . Another mechanism could be the significantly high N protein expression in SARS-CoV-2-infection [49] . N protein was reported to antagonize IFNβ response in SARS-CoV infected cells [50] . IFN synthesis and signaling have also been found to be antagonized by SARS-CoV open reading frame (Orf) 3b and Orf6 [51] . Innate immunity is mainly mediated via interferons (IFN), which are the first line of defense against viral infections by activating macrophages and natural killer (NK) cells. This interferes with the production of viral proteins by presenting antigens to cytotoxic T-cells that can directly bind and destroy the virus-infected host cells, hence preventing severe consequences of the disease [4, 52] . Crosstalk between macrophages, conventional dendritic cells and plasmacytoid dendritic cells are a major cellular pathway for the control of severe and fatal cytopathic virus infection [53] . All three types of interferons (I, II, and III IFNs) share a common signal transducer and activator of transcription 1 (STAT1) that control the expression of several IFN regulated genes [54] . Deletion of any of the signaling components involved in the STAT1 signaling pathway significantly weakens the innate immune response and increases susceptibility to several pathogens, including viruses [55] . STAT1 has also been reported to play a role in adaptive immune processes, especially those seen in SARS-CoV patients with severe infection [55] . MERS-CoV, SARS-CoV and SARS-CoV-2 have been found to encode proteins that contribute to the suppression of IFN by avoiding interferon-stimulated gene (ISG) effector functions and evading antiviral innate immune pathways [52, [56] [57] [58] [59] [60] [61] [62] . This occurs in severe outcome patients that showed lower ISG and immunoglobulin gene expression levels, persistent chemokine levels, and deficient anti-SARS spike antibody production [63, 64] . However, during SARS-CoV-2 infections, levels of IFN-I and IFN-III have also been shown to be low despite sufficient ISG expression, which resulted in a decrease in the innate antiviral response [65, 66] . This low IFN level was accompanied with elevation in proinflammatory cytokines, which culminated in an inadequate antiviral response in a hyperinflammatory setting that can explain the pathogenicity of severe COVID-19 cases [65] . In patients with severe COVID-19 enhanced expression of TNF-α, macrophage inflammatory protein 1-α (MIP-1α), GM-CSF, IL-2, IL-6, MCP3, and IP-10 were detected. Excessive levels of chemokines (CXCL1, IP-10, CXCL5, CCL2/MCP1, CXCL10) were also demonstrated among those patients with severe COVID-19 [13, [67] [68] [69] [70] . The higher the infectiveness of SARS-CoV-2, the lower the levels of IFNs and proinflammatory cytokines/chemokines are, compared to SARS-CoV (52) . SARS-CoV-2 reportedly does not trigger any IFN response, and only significantly activates 5 of the 13 proinflammatory mediators, compared to 11 of the 13 that are activated during SARS-CoV infection. The low degree of innate immune activation can also explain the asymptomatic or mild symptoms in more than 80% of COVID-19 patients [71] . The angiotensin converting enzyme 2 (ACE2) receptor, which is utilized by both SARS-CoV and SARS-CoV-2 for cell entry, is also down regulated by IFN-γ and interleukin-4 (IL-4) [72] . However, unlike IFN-γ that may not be activated, IL-4 acts early during the replication cycle of the virus [72] . Pathogenic levels of immune complexes (ICs) are commonly seen in several disorders, such as serum sickness or viral diseases where IC deposition and excessive inflammatory reactions have been reported [73] . The involvement of ICs in the pathogenesis of severe cases of SARS-CoV-2 infection is supported by the late development of the cytokine storm, especially endothelitis and disseminated microvascular J o u r n a l P r e -p r o o f thrombosis, which affects multiple organs, including the heart, brain, and kidney [74] [75] [76] . Complement proteins bind to erythrocytes that then carry ICs to phagocytes in the liver and the spleen. The role and activation of the complement and coagulation pathways in the setting of SARS-CoV-2 infection are controversial; despite most studies confirming an essential role [77] [78] [79] [80] . Additionally, to activate the innate type-I interferon and IL-6 dependent inflammatory immune responses, complement function was found to modulate and predict immunity, susceptibility, and clinical outcome associated with SARS-CoV-2 infection [81] . For example, the activation of the complement protein, C3, occurs early and contributes to the prothrombotic and proinflammatory states culminating in end organ damage seen in severe cases of COVID-19 [82] . In severe COVID-19 cases, high inflammatory markers were associated with pathologically low levels of C3 and C4 complement components. These low levels were concluded to be a result of excessive activation of the complement pathway with the consequent lung pathology, which resulted in declining C3 level [80] . Lymphocytes T lymphocytes are of high importance for clearing an infectious process that has begun, as their plasma levels correlate with higher survival [83, 84] . Activated killer T cells in response to SARS-CoV-2 have the ability to prevent the spread of the virus from the upper respiratory tract. Hence, the efficiency of this response will determine the fate of the symptom severity, host viral load and transmission rates into the community. Leukopenia and lymphocytopenia have been proposed as a way the virus evade the host immune response [85] [86] [87] . At the time of SARS-CoV infection, CD8 + T cell depletion did not affect viral clearance or replication, whereas CD4 + T cell depletion resulted in pathologic consequences represented by interstitial pneumonitis and delayed clearance of infection from the lungs [18] . This CD4 + T cell depletion and delay in viral clearance was found to be associated with a reduction in the production of cytokines, neutralizing antibodies, and the recruitment of lymphocytes to lung tissue [18] , highlighting the impact of COVID-19 on CD4 + T lymphocyte [79, 88] . Severity, mortality, and dysregulated immunological response towards SARS-CoV-2 induces lymphopenic community-acquired pneumonia (L-CAP), and the presence of lymphopenia and hypercytokinemia indicates poor control of the pathogen [7, 48, 89, 90] . This has also been seen in patients with severe SARS-CoV infections [63, 64] . The low lymphocyte counts have been suggested to be due to the direct cytotoxic action of the virus by preventing cytokine storm and dampening the innate immune responses [90] [91] [92] . Multiple genes involved in apoptosis and P53 signaling were found to be upregulated in patients with COVID-19, which may help explain the development of lymphopenia in these patients [70] . In addition, host factors have also been reported to contribute to the lymphopenic state in severe SARS-CoV-2 infections, as patients requiring intensive care generally have comorbidities such as hypertension, diabetes, cardiovascular and cerebrovascular disease and are commonly older than those with mild disease presentation [7] . Both older age and comorbidities are associated with endothelial dysfunction, which increases the risk of enhanced leukocyte adhesion and extravasation and contributes to lymphopenia seen in severe SARS-CoV-2 infections [93, 94] . During severe SARS-CoV infection, an unusual conversion of B lymphocytes to macrophage-like cells leads to inability of the humoral and cellular component of the immune system to respond in time to neutralize viral infection [95, 96] . This is thought to be driven by the spike protein of SARS virus, and the local hypoxia seen in patients with severe SARS-CoV infection [96] . COVID-19 has led to an exhaustion of effector T cells, negatively affecting their defense against SARS-CoV-2 [97, 98] . This loss of effector T cell function is either due to the increased expression of inhibitory receptors on the surface of T cells due to cytokine activity or a reduction in regulatory T cells [79, 99] . The membrane (M), spike (S), nucleocapsid (N), open reading frame (ORF), mainly ORF3a, and ORF8 proteins, and non-structural proteins (NSP), specifically NSP3 and NSP4, are all proteins targeted by SARS-CoV-2−specific CD8 + and CD4 + T cells. The presence of S-reactive CD4 + T cells was reported in 83% of patients with SARS-CoV-2 infection. Interestingly, the detection of SARS-CoV-2−reactive CD4 + T cells in healthy controls highlighted possible cross-reactive T cell recognition between circulating CoV and SARS-CoV-2 [100] . Studies have demonstrated the presence of SARS-CoV-2-reactive CD4 + T cells in 20% of SARS-CoV-2 seronegative healthy donors in the Netherlands [88] , 34-81% in Germany [101, 102] , 40-60% in the United States of America [100] , 51% in Singapore [103] , and 30% in the United Kingdom [104] . However, a study from the UK did not detect any SARS-CoV-2-reactive T cell responses in unexposed healthy volunteers [105] , and a study from Wuhan failed to detect any spike-specific T cell responses before vaccination on 108 volunteers without pre-exposure to COVID-19 [106] . Humoral response against SARS-CoV-2 has been found to be comparable to other CoVs, SARS-CoV in particular [107] [108] [109] [110] [111] . During SARS-CoV-2 infections, SARS-specific immunoglobulin M (IgM) antibodies were found to appear two weeks (can be as early as 3 to 6 days) after the onset of the infection, last one month before they gradually start to disappear until the end of week 12 [107, [109] [110] [111] (Fig.1) . Immunoglobulin G (IgG) was produced 16 to 18 days after the onset of infection [107, 109] (Fig.1) and to last for a longer but limited time in COVID-19 patients [107] . Overall, the antibody responses for SARS-CoVs do not last for a very long time [112] [113] [114] . SARS-CoV antibodies were not detectable in 91% of tested samples when measured six years after SARS-CoV infection [115] . In SARS-CoV-2 infected patients, detection of both IgM and IgG antibodies are used to identify the stage of the infection. While it is still unclear on how long antibodies to SARS-CoV-2 will be present in the system, a recent study reported the level of anti-SARS-CoV-2 antibodies reduced by half-life during week 4-12 following the start of infection [116] . However, studies have shown that patients can recover from SARS-CoV-2 infection without functional B cells, highlighting that despite the importance of the antibody response, it is not essential [117, 118] . While CoV infections were shown to trigger an antibody response, this usually results in the formation of neutralizing or enhancing antibodies as seen in SARS-CoV and MERS-CoV [119] [120] [121] . Enhancing antibodies and neutralizing antibodies can counteract each other's function [122] . Neutralizing antibodies exert their action via blocking the entry, egress, or the fusion of the virus into the host cell; for example, recognition of spike protein RBD by neutralizing antibodies blocks viral entry [123] . Even though virusspecific antibodies are considered to have antiviral effects and aid in viral clearance, the presence of specific antibodies can enhance viral infections through antibody-dependent enhancement (ADE) [124] . ADE was correlated with an increase in the level of proinflammatory mediators and a decrease in antiinflammatory mediators [125] . In SARS-CoV infection, ADE was facilitated by the engagement of Fc receptors on different immune cells, which facilitated viral entry [126] [127] [128] [129] [130] . Whether the triggered antibodies will have enhancing or neutralizing properties, is proposed to be a function of antibody quality and quantity. Antibodies produced with higher concentrations, and those with higher affinity are less likely to result in ADE [131] . In the setting of ADE, engagement of Fc receptors of immune cells is proposed as an ACE2 independent path of viral entry into host cells [130] (Fig. 2,3) . SARS-COV-2 antigenic mimicry with human tissue has received attention recently. It was found that several host tissue antigens had strong reactions with the SARS-CoV-2 antibodies; this highlighted that not only pulmonary tissue antigens, but several other tissue antigens cross-reacted with SARS-CoV-2 proteins [132, 133] . Specifically, the relationship between SARS-CoV-2 spike, nuclear proteins and autoimmune target proteins highlighted the possibility of an autoimmune reaction against human tissues resulting in the extensive organ, tissue, and cellular damage seen in severe SARS-COV-2 infections, this significant reaction can potentially result in an autoimmune reaction against host proteins and tissues such as pulmonary surfactant proteins, connective tissue, the respiratory, digestive, cardiovascular, and nervous systems [132] [133] [134] [135] . It is worth mentioning that a recent study demonstrated that the risk of developing severe COVID-19 phenotype among patients with autoimmune diseases concluded no difference compared to controls [136] . This can be due to their concomitant use of immune suppressant, which can play a role in modulating cytokines storm in severe COVID-19 cases. The cross-reactivity between CoVs, SARS-CoV, MERS-CoV, and SARS-CoV-2 is unclear. A study of COVID-19 specific humoral immune response has shown that the patient's produced IgM and IgG antibodies, cross-reacted with SARS-CoV, but not with other human CoVs (HCoVs) [137] . If crossreactivity exists between pre-existing antibodies against other CoVs, then cross-reactive ADE is possible where infection with the new SARS-CoV-2 can be enhanced, resulting in a more severe illness or a faster adaptive immune response (Fig. 2, 3) . The higher prevalence of common CoVs in previous years in some regions in the world, might explain the higher pathogenicity of SARS-CoV-2 infection in these affected regions based on the assumption of ADE with pre-existing enhancing antibodies against those common CoVs. For example, the link between early response with higher titers and older age may indicate a priming effect from existing antibodies against other endemic strains [128] . The HCoV circulate continuously, therefore, it is sensible to assume that CoVs antibodies are higher in older people compared to children, including enhancing antibodies [138] . Children infected by SARS-CoV-2 usually have milder presentation of the disease [139] . However, we do not have enough evidence to conclude such role of ADE. This possibility of ADE makes it doubtful that the use of convalescent plasma to treat SARS-CoV-2 infection is of benefit, as theoretically, it can potentially lead to transmission of enhancing antibodies from individuals who recovered from SARS-CoV-2 infection, hence worsening of the disease in recipients. Some studies reported a lack of effectiveness of convalescent plasma and induction of endothelial damage, while clinical studies did not reveal any deleterious effects of using convalescent plasma to treat SARS-CoV-2 infection which makes the possibility of transmission of enhancing antibodies unlikely [122, 140] . The mutations present at the spike level of SARS-CoV-2 variants are of concern and have been categorized as immune escapes. It is possible that these mutations influence ADE antibody elicitation, thus making the human host more or less susceptible to infection and reinfection irrespective of possible cross-reactivity of antibodies. Nevertheless, the issue of enhancing antibodies is of great importance for the development of a vaccine against SARS-CoV-2, which ideally should not aim to induce enhancing antibodies in healthy individuals [130] . To date, studies on currently available vaccines excluded the possibility of vaccine triggering enhancing antibodies. Nevertheless, identifying the SARS-CoV-2 neutralizing, cross-neutralizing, and enhancing epitopes is essential for any vaccine design to minimize the risk of ADE. Epitopes of T cell should also be recognized to outline protective immunity [122, 141] . where non-neutralizing antibodies (non-NAb) bind to SARS-CoV-2 but lack the ability to neutralize it, the NAb-virus complex binds to immune cells via the Fc receptors (FcR) on their surface, this results in marked increase in the release of inflammatory cytokines (cytokine storm) and suppression of anti-inflammatory cytokines. If cross-reactivity with other human coronaviruses (hCoV) exists and triggers high concentration of antibodies with strong affinity this results in an efficient immune response and subside SARS-CoV-2. If cross-reactivity with other hCoV exists but rather triggers low concentration of antibodies with low affinity this results ADE where non-NAb mediate massive increase in inflammatory cytokines which results in severe SARS-CoV-2 infection with cytokine storm as a hallmark. Created with BioRender.com The potential cross-reactivity between seasonal HCoVs, SARS-CoV and MERS-CoV and the pandemic SARS-CoV-2 can have implications on the course of COVID-19 natural infection. While the immunity to some HCoVs such as HCoVOC43 and HCoVHKU1 has been shown to fade within one-year [25, 142] , SARS-CoV and MERS-CoV infections can potentially induce longer-lasting immunity [143, 144] . Memory T-cell responses specific for SARS-CoV have been detected two years after recovery [64] . In general, betacoronaviruses have the potential to trigger immune responses against one another by sharing antigen epitopes for presentation to the immune system via major histocompatibility (MHC) class I receptors [145] . This is why significant titers of cross-reactive antibodies against other betacoronaviruses were detected in sera of SARS-CoV patients [143, 145, 146] . SARS-CoV and HCoV-OC43 infections have been shown to result in cross-reactive antibodies against MERS-CoV and SARS-CoV, respectively [143, 146] . However, some studies reported that antibodies induced by SARS-CoV are unable to crossneutralize MERS-CoV [147] . The strongest cross-reactivity was detected between SARS-CoV-2 and MERS-CoV/SARS-CoV antibodies due to genetic sequence identity [148] . Nevertheless, cross-reactivity of antibodies against SARS-CoV-2 with MERS-CoV, SARS-CoV, HCoV-OC43, and HCoV-HKU is controversial [109, 144, 148, 149] . While some studies demonstrated that the sera of SARS-CoV patients concluded no cross-neutralization against SARS-CoV-2 implicating the lack of cross-protection, others have reported that SARS-CoV-2 antibodies cross-reacted with SARS-CoV [109, 144, 146, 148, 150] . It is unknown whether cross-reactive memory T cells may exacerbate COVID-19 disease or reduce its severity [151] . Increased severity of COVID-19 due to cross reactivity can be due to poor avidities of T cell receptors or abnormal T cell polarization. The concept of cross-reactivity is of great importance however, since taking measures to contain COVID-19 will mean less spread of SARS-CoV-2, it also means less spread of herd immunity to HCoV [152, 153] . The consequence of this is the presence of low levels of cross-immunity between other betacoronaviruses and SARS-CoV-2. Ultimately, the result could be that SARS-CoV-2 will start to fade before a resurgence after some time [145] . It is important to note that cross-reactivity does not necessarily mean cross-protection. Cross-reactivity has also been reported between Dengue virus (DENV) and Zika virus (ZIKV) as their immunological cross-reactivity is expected due to the similarity in their geographic distributions and high sequence homology. While T cell responses may help explain the cross-protection against Zika virus from a previous Dengue virus infection, cross-reactive antibody responses may increase the risk of severe secondary Dengue virus infections [154] . Similarly, it was shown that ADE triggered by a previous CoV infection could increase susceptibility to and the severity of SARS-CoV-2 infection [155, 156] . Thus, the development of cross-reactive T cell responses and cross-reactive antibodies may prevent or promote severe disease, depending on their precise balance upon the secondary infection with a different serotype ( fig. 3 ). Subset alteration of lymphocytes were found to be associated with both clinical characteristics and treatment efficacy of COVID-19 [157] . Specifically, cytotoxic T lymphocytes have been reported to be an independent predictor for COVID-19 severity and treatment efficacy [157] . Cross-reactivity may explain the vital involvement of humoral immune response in vaccine-mediated protection against-and recovery from SARS-CoV infection, which should be accompanied by the development of a neutralizing antibody response [158] [159] [160] [161] . These SARS-CoV-specific neutralizing antibodies developed during convalescence were found to prevent the reinfection with the virus in animal models [162] . A study from the United States reported reinfection in a patient with two distinct strains of the SARS-CoV-2 virus, and suggested that previous exposure to the virus would not result in total immunity [163] . Susceptibility to SARS-CoV-2 can be difficult to define due to largely unknown pre-existing crossprotective immunity in individuals exposed to antigenically related pathogens, including viruses [164] . Reduction in symptom severity and transmission rates of homologous and heterologous HCoVs have been reported in the setting of prior immunity by previous exposure to one HCoV [25, 165, 166] . However, cross-protective immunity with HCoV is not thought to be long-lasting [167] , as repeated infections have been reported in all age groups with both homologous and heterologous HCoVs [25, 26] . This might explain the age-related susceptibility to and increased severity of COVID-19 among the elderly as higher HCoV infection rates are seen in children [166, 168, 169] and reported to be associated with relative protection from SARS-CoV-2 [170] . Despite the scarcity of data regarding the relationship between aging and the host response to virus infection, few studies provided a potential explanation for the increased susceptibility and more severe lung pathology following SARS-CoV infection. Baas et al. reported an age-dependent innate immune response to SARS-CoV in mice. Innate immune response to SARS-CoV was found to differ with the age of the host; older mice respond with a faster and exacerbated innate immune response than younger mice but had a delay in the virus clearance [18, 55, 171, 172] . Animal and human studies are yet to confirm these findings in SARS-CoV-2 infection. Most importantly, a second wave of a biphasic response in aged mice was found to trigger a subset of genes that is responsible for the activation of T lymphocytes leading to severe histopathologic changes in the lungs [18, 55, [171] [172] [173] . Similarly, higher levels of PGD 2 seen in the lungs of older patients with severe respiratory infections have been found to negatively affect the initial innate immune and the later adaptive immune responses [174] . Studies in macaques using SARS-CoV also concluded a more robust innate immune response in older macaques, while IFN-β expression as an anti-inflammatory cytokine was found to be low in older macaques [175] . Despite rare deaths and better prognosis in children younger than 15 years, among the rare severe cases, they were found to have more severe symptoms than those displayed by older children [139] . Higher ACE2 activity in younger children [25, 26, 145, 146, [165] [166] [167] [168] [169] [170] , compared to those older than 13, have been noticed and suggested to explain the more severe symptoms among the younger children [176] . This higher ACE2 activity is not believed to result in severe complications in children as in adults [177] . In addition, SARS-CoV-2 has been shown to be associated with a decreased count of CD4 cells; children have been found to have higher numbers of CD4 cells compared to adults, this may explain their better prognosis [178, 179] . Although rare, severe cases of COVID-19 among children have been reported, SARS-CoV-2 in these cases has been associated with triggering Kawasaki disease and toxic shock syndrome [141, 180] . Over the past 18 months, the SARS-CoV-2 virus has started to acquire mutations, approximately two single-letter mutations per month in its genome, resulting in new variants. Several significant, more fit to survive, variants have emerged. As SARS-CoV-2 began to spread rapidly, the number of immune individuals was insignificant, so the variant with more efficient transmission prevailed and gained an advantage over other circulating strains. With the greater spread of infection, and as the number of immune individuals increased, the virus started to gain benefits from evading naturally induced immunity where it can potentially reinfect the same individual, so variants with better ability to evade naturally induced immunity have better chances for survival in the new host. Those variants have been labelled by the World Health Organization as variants of concern (VoC). With the initiation of a global vaccination program, those VoCs can potentially threaten to elude vaccine-induced immunity, which may jeopardize vaccination efficacy. Eventually, when much of the susceptible population is vaccinated with effective vaccines, the variant better suited for survival in the new host will be the one with a better ability to evade the vaccine-induced immunity. However, variants that are able to evade vaccine-induced immunity have not yet been reported and may not necessarily emerge [181] . One of the first identified variants includes the D614G mutation in the Spike (S) protein modification [182, 183] . This mutation targets amino acid 614, which is located outside the receptor binding domain (RBD) and is known to enhance viral infectivity by shifting the S protein conformation towards ACE-2 binding fusion state to allow viral entry and replication [182, 184, 185] . Another variant that emerged in North Jutland, Denmark but with very limited spread is the Cluster 5 SARS-CoV-2 variant which was related to infection spread in mink farms and was transmitted to humans. This variant has been shown to have a combination of mutations that have not been observed before. To date, following extensive investigation and surveillance, Danish authorities have identified only 12 human cases of the Cluster 5 variant up to September 2020, and it does not appear to have spread widely. 1 have been declared as variants of concern (VoC) by the WHO, as these variants share common characteristics, including escape mutations and impact neutralization efficacy. While variant B.1.1.7 has been shown to be more sensitive to neutralization, it does not appear to evade the immune system [187, 188] . Mutations present in the B.1.351 and P.1 variant (K417T, E484K, and N501Y) are highly concerning, since they have shown to compromise neutralization that was generated by a previous infection or vaccination and may increase viral infectivity and fitness [189] [190] [191] . Two other SARS-CoV-2 sequences belonging to the B.1.1.207 lineage have been reported recently in Nigeria. These sequences have been shown to share one non-synonymous mutation in the spike protein (P681H) in common with the UK B.1.1.7 variant; none of the other 22 unique mutations of the B.1.1.7 lineage was detected in these sequences. The P681H mutation occurred at the highly variable region near the S1/S2 furin cleavage site [192] . Among the many concerns of the emergence of new variants, the ability to evade natural or vaccineinduced immunity is of most concern. To be able to evade natural or vaccine-induced immunity, the virus will need to accumulate several mutations in the spike protein to be able to overcome the polyclonal immune response to several parts of the spike protein triggered by vaccines or natural infection with SARS-CoV-2. The most dominant mutation worldwide is presented as SARS-CoV-2 with the D614G variant. In vitro studies indicate that this mutation confers greater infectivity, while molecular epidemiology correlates it with an increase in transmissibility with no evidence to date for increased virulence. This variant was shown to be highly sensitive to natural or vaccine-derived neutralizing antibodies, which can be due to the effect of this mutation on the S protein [184, 193] , so this variant is unexpected to threaten the antibody-mediated immunity produced in response to the original D614 S protein. The Denmark cluster 5 variant has been suggested to have the potential to reduce virus neutralization in humans and consequently decrease the duration of immunity triggered after natural infection or vaccination. Mutations that target regions located outside the RBD have not been associated with increased severity; however, they have been reported to result in increased infectivity and transmissibility [194] . Several studies concluded that SARS-CoV-2 with the D614G variant is highly sensitive to neutralizing antibodies, natural or vaccine-derived, which can be due to the effect of this mutation on the S protein [184, 193] . The RBD is immunodominant and accounts for 90% of serum neutralizing activity [195] . Mutations in the RBD have been reportedly associated with a higher potential of evading the immune system. The RBD variant N439K has been found to result in enhanced RBD affinity for ACE2 and higher viral load, it also was concluded to be able to evade antibody-dependent immunity, but no change in disease severity was associated with this variant. B.1.1.7 variant (Alpha variant) that first emerged in the UK before it spreads to several other countries has been shown to be associated with an increased fatality compared with other variants [192] . For the B.1.351variant (Beta variant) of SARS-CoV-2 that has emerged in South Africa, there is no evidence of any impact on disease severity. There is some evidence though, that linked E484K, one of the spike protein mutations, with an effect on neutralization by some polyclonal and monoclonal antibodies [196, 197] . At the time of P.1 variant (Gamma variant) emergence, Manaus, Brazil was expected to have surpassed the theoretical herd immunity threshold (67%) [198] . It was reported that this variant might influence the transmissibility and antigenic profile of SARS-CoV-2, which may affect the ability of antibodies generated through previous natural infection or through vaccination to recognize and neutralize the virus; a study reported that this variant might have the ability to elude the human immune response that was triggered by the previous variant. Although sequencing of the variants that emerged in Nigeria (B.1.1.207) has shown no signs of increased virulence, it is still unknown whether these variants have any impact on the transmission or disease severity of SARS-CoV-2 in Nigeria [192] . Table 1 displays the main variants of SARS-CoV-2 and the consequent change in transmissibility, virulence, and antigenicity. Further studies must investigate the impact of the new variants of concern on reinfection, resurgence, cross-reactivity, and ADE. In-vitro evidence demonstrates that the presence of the E484K mutation reduces the neutralization of multiclonal antibodies in convalescent sera [190] . It is noteworthy to mention that lack of neutralization does not essentially mean lack of protection from disease antibodies; although important, they are not the only immune component in the fight against SARS-CoV-2. The marginal amount of antibodies can still protect from SARS-CoV-2, in fact, the possibility of reinfection can also provide an immunity boost that has been seen in other viral infections such as rubella, where reinfection was shown to be clinically insignificant, patients were noninfectious and boosted immunity against the virus [199] . It is also of high importance to take T cell responses into consideration when talking about immunity towards these new variants. A recent study has shown that, compared to antibodies [200] , T cells can be more resilient to threats from emerging variants. This proposed resilience is reportedly due to the fact T cells generated in response to SARS-CoV-2 were found to target at least 15 to 20 different fragments of coronavirus proteins which hinders the virus's ability to escape cell recognition. In fact, a recent study reported that most T-cell responses are unlikely to be altered by the mutations in emerging variants[201]. Understanding the mechanisms associated to the innate immunity provides us a better understanding of the pathogenesis of the disease and a promising therapeutic treatment. Despite the increasing number of studies on the immunologic aspects of SARS-CoV-2 infection, more studies are still needed to adequately help predict the progression of the infection into asymptomatic or critical phenotypes. An early inadequate IFN antiviral response, and a later overproduction of proinflammatory cytokines, ADE, infection of innate immune cells, recruitment of uninfected cells from the circulation to the primary site of infection, and/or the consequent massive immune reactions induce hyperinflammation, may play a vital role in the progression to life-threatening complications seen in severe COVID-19 cases. The roles which SARS-CoV-2-specific T cell phenotypes, autophagy, ADE, and cross-reactivity may play against SARS-CoV-2 needs to be further investigated. Currently, there is also a lack of details of the roles of the hyperactivation of the immune system to the lung and the systemic damage that can lead to patients' death [142, 202] . The increased virus infections provide the basis for SARS-CoV-2 to mutate and better evade host immunity, which has led to new variants showing higher transmissibility rates. Any future global immunization program will need to cover immunity against new coronavirus variants to ensure the effectiveness of these vaccination programs. Further research must be conducted the investigate the transmission, virulence, and antigenicity of the VoC's, and its association to the COVID-19 heterogeneous phenotypes. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding A systematic review of antibody mediated immunity to coronaviruses: antibody kinetics, correlates of protection, and association of antibody responses with severity of disease. medRxiv T cell responses to viral infections -opportunities for Peptide vaccination. Front Immunol Transmission of 2019-nCoV Infection from an Asymptomatic Contact in Germany Receptor Recognition by the Novel Coronavirus from Wuhan: an Analysis Based on Decade-Long Structural Studies of SARS Coronavirus Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China Clinical Characteristics of Coronavirus Disease 2019 in China Coronavirus Endoribonuclease and Deubiquitinating Interferon Antagonists Differentially Modulate the Host Response during Replication in Macrophages Severe acute respiratory syndrome coronavirus M protein inhibits type I interferon production by impeding the formation of TRAF3.TANK.TBK1/IKKepsilon complex COVID 19: a clue from innate immunity COVID-19, cytokines and immunosuppression: what can we learn from severe acute respiratory syndrome? COVID-19: consider cytokine storm syndromes and immunosuppression COVID-19 cytokine storm: the interplay between inflammation and coagulation The pathogenesis and treatment of the `Cytokine Storm' in COVID-19 On the Alert for Cytokine Storm: Immunopathology in COVID-19. Arthritis Rheumatol Immunity and immunopathology to viruses: what decides the outcome? Cellular immune responses to severe acute respiratory syndrome coronavirus (SARS-CoV) infection in senescent BALB/c mice: CD4+ T cells are important in control of SARS-CoV infection Resolution of primary severe acute respiratory syndrome-associated coronavirus infection requires Stat1 MyD88 is required for protection from lethal infection with a mouse-adapted SARS-CoV Expression profile of immune response genes in patients with Severe Acute Respiratory Syndrome from interferons to cytokines Extraordinary GU-rich single-strand RNA identified from SARS coronavirus contributes an excessive innate immune response. Microbes Infect Human Influenza Virus Infections. Semin Respir Crit Care Med The time course of the immune response to experimental coronavirus infection of man Human Coronavirus NL63 Molecular Epidemiology and Evolutionary Patterns in Rural Coastal Kenya Natural SIV hosts: showing AIDS the door Optimal immune responses: immunocompetence revisited Innate Immune Responses to Highly Pathogenic Coronaviruses and Other Significant Respiratory Viral Infections. Front Immunol Targeting the NLRP3 Inflammasome in Severe COVID-19 Measles virus V protein inhibits NLRP3 inflammasome-mediated interleukin-1β secretion Coronavirus membrane-associated papain-like proteases induce autophagy through interacting with Beclin1 to negatively regulate antiviral innate immunity NLRP3 Inflammasome-A Key Player in Antiviral Responses. Front Immunol The RNA-and TRIM25-Binding Domains of Influenza Virus NS1 Protein Are Essential for Suppression of NLRP3 Inflammasome-Mediated Interleukin-1β Secretion Sendai Virus V Protein Inhibits the Secretion of Interleukin-1β by Preventing NLRP3 Inflammasome Assembly An NLRP3 inflammasome-triggered cytokine storm contributes to Streptococcal toxic shock-like syndrome (STSLS). PLoS pathogens NLRP3 Inflammasome Regulates Development of Systemic Inflammatory Response and Compensatory Anti-Inflammatory Response Syndromes in Mice With Acute Pancreatitis Into the eye of the cytokine storm Cytokine storm and sepsis disease pathogenesis COVID-19, ECMO, and lymphopenia: a word of caution. The Lancet Respiratory Medicine Immune response to SARS-CoV-2 and mechanisms of immunopathological changes in COVID-19 Mannose-binding lectin in severe acute respiratory syndrome coronavirus infection Functional polymorphisms of the CCL2 and MBL genes cumulatively increase susceptibility to severe acute respiratory syndrome coronavirus infection Association between mannose-binding lectin gene polymorphisms and susceptibility to severe acute respiratory syndrome coronavirus infection Mannose-binding lectin is associated with thrombosis and coagulopathy in critically ill COVID-19 patients. Thrombosis and haemostasis Induction of pro-inflammatory cytokines (IL-1 and IL-6) and lung inflammation by Coronavirus-19 (COVI-19 or SARS-CoV-2): anti-inflammatory strategies The trinity of COVID-19: immunity, inflammation and intervention Clinical features of patients infected with 2019 novel coronavirus in Wuhan Comparative replication and immune activation profiles of SARS-CoV-2 and SARS-CoV in human lungs: an ex vivo study with implications for the pathogenesis of COVID-19 The severe acute respiratory syndrome coronavirus nucleocapsid inhibits type I interferon production by interfering with TRIM25-mediated RIG-I ubiquitination Severe acute respiratory syndrome coronavirus open reading frame (ORF) 3b, ORF 6, and nucleocapsid proteins function as interferon antagonists SARS coronavirus pathogenesis: host innate immune responses and viral antagonism of interferon Type I IFN-mediated protection of macrophages and dendritic cells secures control of murine coronavirus infection Transcriptional responses to polypeptide ligands: the JAK-STAT pathway SARS-CoV pathogenesis is regulated by a STAT1 dependent but a type I, II and III interferon receptor independent mechanism SARS coronavirus and innate immunity Innate immunity during SARS-CoV-2: evasion strategies and activation trigger hypoxia and vascular damage Regulation of IRF-3-dependent innate immunity by the papain-like protease domain of the severe acute respiratory syndrome coronavirus Interferon and cytokine responses to SARS-coronavirus infection Coronaviruses post-SARS: update on replication and pathogenesis Human cell tropism and innate immune system interactions of human respiratory coronavirus EMC compared to those of severe acute respiratory syndrome coronavirus The structural and accessory proteins M, ORF 4a, ORF 4b, and ORF 5 of Middle East respiratory syndrome coronavirus (MERS-CoV) are potent interferon antagonists Interferon-mediated immunopathological events are associated with atypical innate and adaptive immune responses in patients with severe acute respiratory syndrome Human immunopathogenesis of severe acute respiratory syndrome (SARS) Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19 Overview of Immune Response During SARS-CoV-2 Infection: Lessons From the Past. Front Immunol Heightened Innate Immune Responses in the Respiratory Tract of COVID-19 Patients The use of anti-inflammatory drugs in the treatment of people with severe coronavirus disease 2019 (COVID-19): The Perspectives of clinical immunologists from China The cytokine storm in COVID-19: An overview of the involvement of the chemokine/chemokine-receptor system Transcriptomic characteristics of bronchoalveolar lavage fluid and peripheral blood mononuclear cells in COVID-19 patients Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: summary of a report of 72 314 cases from the Chinese Center for Disease Control and Prevention Interferon-gamma and interleukin-4 downregulate expression of the SARS coronavirus receptor ACE2 in Vero E6 cells Immune complex disease COVID-19: Immunology and treatment options Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study A plea for the pathogenic role of immune complexes in severe Covid-19 C3 and ACE1 polymorphisms are more important confounders in the spread and outcome of COVID-19 in comparison with ABO polymorphism Clinical Features and Treatment of 2019-nCov Pneumonia Patients in Wuhan: Report of A Couple Cases Dysregulation of Immune Response in Patients With Coronavirus Decreased complement C3 levels are associated with poor prognosis in patients with COVID-19: A retrospective cohort study Identification of Immune complement function as a determinant of adverse SARS-CoV-2 infection outcome. medRxiv Microvascular COVID-19 lung vessels obstructive thromboinflammatory syndrome (MicroCLOTS): an atypical acute respiratory distress syndrome working hypothesis T cell-mediated immune response to respiratory coronaviruses T cell responses to whole SARS coronavirus in humans Haematological manifestations in patients with severe acute respiratory syndrome: retrospective analysis Significant changes of peripheral T lymphocyte subsets in patients with severe acute respiratory syndrome. The Journal of infectious diseases Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia Phenotype and kinetics of SARS-CoV-2-specific T cells in COVID-19 patients with acute respiratory distress syndrome Lymphopenic community-acquired pneumonia is associated with a dysregulated immune response and increased severity and mortality Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study Adaptive immune cells temper initial innate responses Shared Features of Endothelial Dysfunction between Sepsis and Its Preceding Risk Factors (Aging and Chronic Disease) Simultaneous Depression of Immunological Synapse and Endothelial Injury is Associated with Organ Dysfunction in Community-Acquired Pneumonia The clinical pathology of severe acute respiratory syndrome (SARS): a report from China SARS spike protein induces phenotypic conversion of human B cells to macrophage-like cells Elevated exhaustion levels and reduced functional diversity of T cells in peripheral blood may predict severe progression in COVID-19 patients Reduction and Functional Exhaustion of T Cells in Patients With Coronavirus Disease 2019 (COVID-19). Front Immunol CoViD-19 Immunopathology and Immunotherapy Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19 SARS-CoV-2-derived peptides define heterologous and COVID-19-induced T cell recognition SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls Single-Cell Transcriptomic Analysis of SARS-CoV-2 Reactive CD4 (+) T Cells. Ssrn Broad and strong memory CD4(+) and CD8(+) T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19 Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: a dose-escalation, open-label, non-randomised, first-in-human trial Longitudinal Change of Severe Acute Respiratory Syndrome Coronavirus 2 Antibodies in Patients with Coronavirus Disease Profile of specific antibodies to the SARS-associated coronavirus Profiling Early Humoral Response to Diagnose Novel Coronavirus Disease (COVID-19) Antibody Responses to SARS-CoV-2 in Patients With Novel Coronavirus Disease Viral kinetics and antibody responses in patients with COVID-19 Dendritic-cell interactions with HIV: infection and viral dissemination Antibody Response and Disease Severity in Healthcare Worker MERS Survivors. Emerg Infect Dis Two-year prospective study of the humoral immune response of patients with severe acute respiratory syndrome Lack of peripheral memory B cell responses in recovered patients with severe acute respiratory syndrome: a six-year follow-up study High seroprevalence but short-lived immune response to SARS-CoV-2 infection in Paris A possible role for B cells in COVID-19? Lesson from patients with agammaglobulinemia Two X-linked agammaglobulinemia patients develop pneumonia as COVID-19 manifestation but recover Immunodominant SARS Coronavirus Epitopes in Humans Elicited both Enhancing and Neutralizing Effects on Infection in Non-human Primates Molecular Mechanism for Antibody-Dependent Enhancement of Coronavirus Entry Evasion of antibody neutralization in emerging severe acute respiratory syndrome coronaviruses Treatment of 5 Critically Ill Patients With COVID-19 With Convalescent Plasma The spike protein of SARS-CoV--a target for vaccine and therapeutic development Antibody-dependent enhancement of virus infection and disease Prior immunization with severe acute respiratory syndrome (SARS)-associated coronavirus (SARS-CoV) nucleocapsid protein causes severe pneumonia in mice infected with SARS-CoV Microbiologic characteristics, serologic responses, and clinical manifestations in severe acute respiratory syndrome Clinical progression and viral load in a community outbreak of coronavirusassociated SARS pneumonia: a prospective study Neutralizing antibody response and SARS severity Antibody-dependent SARS coronavirus infection is mediated by antibodies against spike proteins Anti-severe acute respiratory syndrome coronavirus spike antibodies trigger infection of human immune cells via a pH-and cysteine protease-independent FcγR pathway Structural insights into the mechanisms of antibody-mediated neutralization of flavivirus infection: implications for vaccine development Autoinflammatory and autoimmune conditions at the crossroad of COVID-19 Covid-19 and autoimmunity Potential antigenic cross-reactivity between SARS-CoV-2 and human tissue with a possible link to an increase in autoimmune diseases On the molecular determinants of the SARS-CoV-2 attack The risk for severe COVID 19 in patients with autoimmune and/or inflammatory diseases: First wave lessons Autophagy and viral neurovirulence Human coronavirus circulation in the United States 2014-2017 SARS-CoV-2 Infection in Children Safety Update: COVID-19 Convalescent Plasma in 20,000 Hospitalized Patients Features of COVID-19 post-infectious cytokine release syndrome in children presenting to the emergency department Cross-reactive antibodies in convalescent SARS patients' sera against the emerging novel human coronavirus EMC (2012) by both immunofluorescent and neutralizing antibody tests Lack of cross-neutralization by SARS patient sera towards SARS-CoV-2. Emerg Microbes Infect Projecting the transmission dynamics of SARS-CoV-2 through the postpandemic period An Outbreak of Human Coronavirus OC43 Infection and Serological Crossreactivity with SARS Coronavirus Antibodies induced by receptor-binding domain in spike protein of SARS-CoV do not cross-neutralize the novel human coronavirus hCoV-EMC Serologic cross-reactivity of SARS-CoV-2 with endemic and seasonal Betacoronaviruses. medRxiv Preexisting and de novo humoral immunity to SARS-CoV-2 in humans Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV Cross-reactive memory T cells and herd immunity to SARS-CoV-2 Preexisting and de novo humoral immunity to SARS-CoV-2 in humans Herd immunity -estimating the level required to halt the COVID-19 epidemics in affected countries Two Is Better Than One: Evidence for T-Cell Cross-Protection Between Dengue and Zika and Implications on Vaccine Design. Front Immunol Is COVID-19 receiving ADE from other coronaviruses? Microbes Infect Understanding SARS-CoV-2-Mediated Inflammatory Responses: From Mechanisms to Potential Therapeutic Tools Characteristics of Peripheral Lymphocyte Subset Alteration in COVID-19 Pneumonia A DNA vaccine induces SARS coronavirus neutralization and protective immunity in mice Highly infectious SARS-CoV pseudotyped virus reveals the cell tropism and its correlation with receptor expression Serology of severe acute respiratory syndrome: implications for surveillance and outcome S protein of severe acute respiratory syndrome-associated coronavirus mediates entry into hepatoma cell lines and is targeted by neutralizing antibodies in infected patients The Immunobiology of SARS* Genomic evidence for reinfection with SARS-CoV-2: a case study. The Lancet Infectious Diseases Estimating age-specific cumulative incidence for the 2009 influenza pandemic: a meta-analysis of A(H1N1)pdm09 serological studies from 19 countries. Influenza Other Respir Viruses Seasonality and immunity to laboratory-confirmed seasonal coronaviruses (HCoV-NL63, HCoV-OC43, and HCoV-229E): results from the Flu Watch cohort study The dominance of human coronavirus OC43 and NL63 infections in infants Coronavirus Occurrence and Transmission Over 8 Years in the HIVE Cohort of Households in Michigan Human coronavirus NL63 and 229E seroconversion in children Human Coronavirus Infections in Israel: Epidemiology, Clinical Symptoms and Summer Seasonality of HCoV-HKU1. Viruses Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection in Children and Adolescents: A Systematic Review Genomic analysis reveals age-dependent innate immune responses to severe acute respiratory syndrome coronavirus Early upregulation of acute respiratory distress syndrome-associated cytokines promotes lethal disease in an aged-mouse model of severe acute respiratory syndrome coronavirus infection Participation of both host and virus factors in induction of severe acute respiratory syndrome (SARS) in F344 rats infected with SARS coronavirus Critical role of phospholipase A2 group IID in age-related susceptibility to severe acute respiratory syndrome-CoV infection Exacerbated innate host response to SARS-CoV in aged non-human primates Serum angiotensin-converting enzyme in healthy and sarcoidotic children: comparison with the reference interval for adults Serum angiotensin-converting enzyme activity in normal children and in those with sarcoidosis Reference ranges for lymphocyte subsets in pediatric patients Cellular immunologic parameters related to age, gender, and stage in lung cancer patients Kawasaki-like multisystem inflammatory syndrome in children during the covid-19 pandemic in Paris, France: prospective observational study Assessing the human immune response to SARS-CoV-2 variants Tracking changes in SARS-CoV-2 Spike: evidence that D614G increases infectivity of the COVID-19 virus Evaluating the Effects of SARS-CoV-2 Spike Mutation D614G on Transmissibility and Pathogenicity Structural and functional analysis of the D614G SARS-CoV-2 spike protein variant Spike mutation D614G alters SARS-CoV-2 fitness SARS-CoV-2 Delta VOC in Scotland: demographics, risk of hospital admission, and vaccine effectiveness. The Lancet Neutralization of SARS-CoV-2 spike 69/70 deletion, E484K and N501Y variants by BNT162b2 vaccine-elicited sera Impact of B. 1.1. 7 variant mutations on antibody recognition of linear SARS-CoV-2 epitopes. medRxiv SARS-CoV-2 501Y. V2 escapes neutralization by South African COVID-19 donor plasma. Nature medicine Comprehensive mapping of mutations to the SARS-CoV-2 receptor-binding domain that affect recognition by polyclonal human serum antibodies Circulating SARS-CoV-2 spike N439K variants maintain fitness while evading antibody-mediated immunity D614G spike mutation increases SARS CoV-2 susceptibility to neutralization. Cell host & microbe SARS-CoV-2 D614G variant exhibits efficient replication ex vivo and transmission in vivo Mapping neutralizing and immunodominant sites on the SARS-CoV-2 spike receptor-binding domain by structure-guided high-resolution serology Escape from neutralizing antibodies by SARS-CoV-2 spike protein variants. Elife Spike E484K mutation in the first SARS-CoV-2 reinfection case confirmed in Brazil Three-quarters attack rate of SARS-CoV-2 in the Brazilian Amazon during a largely unmitigated epidemic Comprehensive analysis of T cell immunodominance and immunoprevalence of SARS-CoV-2 epitopes in COVID-19 cases Multiple host barriers restrict poliovirus trafficking in mice The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.