Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Nov 1.
Published in final edited form as: J Immunol. 2020 Sep 4;205(9):2342–2350. doi: 10.4049/jimmunol.2000839

Antibody Responses to SARS-CoV-2: Let’s stick to known knowns

Nicole Baumgarth 1, Janko Nikolich-Žugich 2,3, F Eun-Hyung Lee 4,5, Deepta Bhattacharya 2,*
PMCID: PMC7578055  NIHMSID: NIHMS1621275  PMID: 32887754

Abstract

The scale of the SARS-CoV-2 pandemic has thrust immunology into the public spotlight in unprecedented ways. In this article, which is part opinion piece and part review, we argue that the normal cadence by which we discuss science with our colleagues failed to properly convey likelihoods of the immune response to SARS-CoV-2 to the public and the media. As a result, biologically implausible outcomes were given equal weight as the principles set by decades of viral immunology. Unsurprisingly, questionable results and alarmist news media articles have filled the void. We suggest an emphasis on setting expectations based on prior findings, while avoiding the overused approach of assuming nothing. After reviewing antibody-mediated immunity after coronavirus and other acute viral infections, we posit that, with few exceptions, the development of protective humoral immunity of more than a year is the norm. Immunity to SARS-CoV-2 is likely to follow the same pattern.

Introduction

The SARS-CoV-2 pandemic has thrust immunology into the public spotlight in unprecedented ways and discussions of antibody testing have become commonplace in the public press, prompting us to write this brief review to put our understanding of humoral immunity to infections in general into the context of infection with this newly emerged virus. As of the writing of this article, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected close to 18 million people and killed over 675,000. The magnitude of this global pandemic is unprecedented in our lifetime. As a result, this virus has received unprecedented attention from scientists and the public. As of this writing, nearly 30,000 Pubmed-indexed articles have already been published about SARS-CoV-2 and COVID-19, the disease it causes. Given that the virus was first reported in December 2019 (1, 2), the pace of investigation and publications makes SARS-CoV-2 the most-studied virus in history. Navigating, filtering, and communicating this enormous amount of information to the public and media has fallen to journal editors and scientists from a variety of diverse disciplines. These are tasks for which few of us were prepared. The implications of what is said and how it is interpreted and publicly scrutinized are greater than anything we have faced to date, and with good fortune, will ever face again.

Communicating with the public, decision-makers, and media

A common refrain in publications, grant applications and interviews is, “Process X is poorly understood.” Science is ultimately a process of reducing uncertainty. Absolute certainty is rarely possible, so expressions of a nuanced approach to addressing a particular question, and revealing a lack of detailed knowledge, are perceived by other scientists as markers of mature, high-integrity work and as providing a sound rationale for studying the details of a problem in depth. Yet, in the context of the COVID-19 pandemic, such statements are often misleading and can be outright harmful. In science, prior knowledge usually sets a range of expectations around which uncertainty is narrowly centered. As scientists, we implicitly understand the nuance and meaning of such statements. Yet at the beginning of the crisis when many, including some of the authors of this perspective and review, were called into interviews with the media, the unintended effects of our standard ways of communicating with other scientists became clear.

It is true that the kinetics of the immune response to SARS-CoV-2, the immunophenotypes of protective innate and adaptive cells, and other key details are poorly understood. Yet, few immunologists would consider it likely that infected subjects would develop no immunity after viral clearance. It is also true that the precise duration of immunity after infection is unknown, but it is unlikely that it lasts only for a few weeks. As we expressed uncertainty, per our normal convention, highly unlikely outcomes were often portrayed by the media as equally likely to those we expected, based on the experience we gained from other viral infections. Public health organizations including the Centers for Disease Control and Prevention (CDC) and the World Health Organization (WHO) have made similar problematic ‘absence of evidence’ statements regarding facemasks, immunity after infection, and the efficacy of testing asymptomatic individuals on college campuses (35). These announcements have had, and still have real-world detrimental consequences on efforts to contain community transmission. They may also engender unwarranted skepticism and hesitancy regarding vaccines and therapeutics, once such options emerge.

Having learned from some of our own misadventures in clearly communicating our thinking, we suggest subtle but meaningful adjustments. Consider the statement, “We don’t know how long immunity will last.” As a comparison, consider the alternative statement, “We don’t know for sure how long immunity will last, but if it is anything like the first SARS coronavirus, it will last for at least several years.” Both statements are factually correct, but the impact on the public is markedly different. We have taken to favor the latter approach. However, to successfully employ this strategy, a critical assessment of the primary data and antecedent literature is essential to form the basis of our answers. What is the evidence that immunity and resistance to subsequent infection are elicited by SARS-CoV-2 infections, and how long is immunity likely to last? How can we apply what we have learned about antibody responses to the interpretation of diagnostic and serological tests? How likely is it that vaccines can protect the general public, as well as vulnerable populations? These questions arise often in daily discourse in contexts we will discuss below. We identify relevant examples in the literature that can provide the basis for setting expectations and answering these questions.

Defining ‘immunity’

To begin, a consistent definition of ‘immunity’ is needed. For the purposes of this article, the operational and objective definition of immunity is a state that increases the inoculum required to cause infection, disease, and transmission. Thus, a loss of immunity implies equal susceptibility to acute viral infections as if one were immunologically naïve. Though in this review we focus on humoral immunity with an emphasis on long-lived plasma cell-derived serum antibodies, many cell types contribute to resistance and immunological memory. Subsets of both memory B and T cells establish local residence at the sites of infection, while others circulate to patrol for distal exposures (6, 7). Natural killer cells form a version of cytokine-competent memory cells (8, 9), and myeloid lineages can maintain an epigenetically primed state for long periods of time (10). The collective sum of these and other components is thus what comprises immunity.

Initial confusion about immunity and re-infection by SARS-CoV-2

During the early stages of the pandemic, reports began to surface of patients testing positive for viral RNA, then negative, and then positive again in relatively short succession (11). The largest number of such subjects exhibiting such a pattern was reported by the South Korean Centers for Disease Control and Prevention (KCDC). A subset of such patients anecdotally reported either continued or new symptoms at the time of the last PCR positive result. Though neither the KCDC, nor many scientists interviewed, considered it likely that these patients had been re-infected in such a short period, this nuance was lost in the news media. Then the WHO issued the following announcement: “there is currently no evidence that people who have recovered from COVID-19 and have antibodies are protected from a second infection.” Unfortunately, these agnotological statements can provide fuel for vaccine skeptics.

Meanwhile, scientists at the KCDC and around the world accumulated evidence that re-infection did not explain these observations, and immunity was almost certain to ensue clearance of the primary infection. Extensive studies by the KCDC demonstrated that infectious virus was undetectable in patients who had tested PCR positive after a negative result (11). Contact tracing of relatives following the return of the patients to their homes revealed not a single instance of transmission (11). These data suggest a few possible explanations for the unusual PCR patterns. First, the second PCR result represents a false negative. Indeed, false negative results can approach 30% of such tests, so mathematically it would be expected that these types of patterns would emerge with some frequency (12). In this case, the patient presumably had not yet cleared the infection in the first place, explaining a subsequent positive result. A corollary to this interpretation is that viral replication may still have been ongoing, but the presence of neutralizing antibodies prevented subsequent culture and community transmission. Second, the last PCR assay might have simply detected viral genomic remnants or non-infectious particles released from dying cells rather than bona fide infectious virus. Consistent with this interpretation, a similar recent study demonstrated that RNA fragments, rather than intact viral genomes were recovered through sequencing in such patients, while culturable virus was not retreivable, nor could virus transmission be detected (13).

Immunity and resistance to re-infection

Concomitantly, several groups reported clear evidence of immunity after SARS-CoV-2 infection and clearance. Seroconversion and neutralizing antibody production, which is the best correlate of protection for many infections and almost all vaccines (14), were observed in nearly all convalescent patients with confirmed COVID-19 (15). For most acute viral infections and following vaccination, the presence of neutralizing antibodies (nAb) is a clear functional correlate of immunity (14), and almost certainly provides at least partial resistance to subsequent infections. Indeed, the first convalescent plasma passive transfer studies in hospitalized patients were also reported around this time (16, 17). Though these studies were small and did not have placebo groups, viral loads were sharply reduced and patient outcomes improved shortly after the plasma transfers. Subsequent convalescent plasma studies in other institutions have continued to suggest a benefit, especially if administered early (18, 19). Another piece of evidence came from experimental infection of macaques. Two weeks after clearance of the first infection, these macaques became fully resistant to high-dose re-challenge with SARS-CoV-2 (20, 21). Collectively, these studies demonstrated what one would have expected, namely that clearance of an acute viral infection leaves one immune.

It is entirely possible that some individuals do not seroconvert after infection, at least to certain viral antigens, particularly if they rapidly cleared a localized upper respiratory tract infection. Perhaps in these cases, the durability of immunity is brief and susceptibility to reinfection is possible. However, the percentages of individuals not seroconverting, claimed by some studies, seem unlikely, ranging from 10–80% (2224), often relying on assays with unclear sensitivities and/or indirect estimates by including individuals who did not have PCR-confirmed infections. Other rigorous studies have shown that seroconversion is almost universal after confirmed infections, even when the disease is mild (15, 2528). Given the evidence, a statement of, “there is no data that COVID-19 recovered patients who develop antibodies can be re-infected” would have conveyed a message of assurance rather than hysteria, and would be entirely consistent with the accumulated evidence.

Challenges with antibody testing and data interpretation for SARS-CoV2

Although the original WHO statement on antibodies and reinfection was off-target in our opinion, a legitimate concern is the degree to which a positive antibody test indicates that one is immune. Beginning in March of 2020, the market became flooded with point-of-care serological tests to define prior SARS-CoV-2 exposure and, presumably, immunity. These products were allowed to be sold under emergency use authorizations by the Food and Drug Administration (FDA), a mechanism employed in response to the initial shortage of RT-PCR tests for viral RNA. Yet, many of these tests performed poorly, reaching false positive rates that approached the true seroprevalence of the target population. Independent validation at the University of California, San Francisco, revealed that the majority of these assays were unreliable, with poor specificity and sensitivity compared to conventional ELISAs (29). The FDA has subsequently removed many of these products in the U.S. Unfortunately, the distinction has been lost between these faulty point-of-care lateral flow assays and reliable standard serological ELISAs, some of which perform exceptionally well and correlate uniformly with virus neutralization titers (26, 27, 30). Such doubt has been amplified in the news media and risks depriving infectious disease epidemiologists of a valuable tool in monitoring community transmission, and also physicians treating COVID-19 subjects of convalescent plasma, proven to contain protective anti-SARS-CoV-2 antibodies.

One of the intended uses of many of the point-of-care tests was to distinguish the timing of infection based on antibody isotype. As primary B cell responses often begin with secretion of IgM, it was assumed that production of this antibody isotype would demonstrate recent, and potentially ongoing infection and virus shedding. Conversely, IgG would represent later phases of infection and convalescence. This interpretation has practical consequences, as some employers, including the Banner-University of Arizona Medical Center-Tucson, require quarantine of employees who test positive for IgM, but dispense with this requirement if SARS-CoV-2-specific IgG is detected.

Fundamentals of B cell responses to acute viral infections

Extrafollicular and germinal center-derived antibody responses to infections shape the serum antibody titers

Despite the broad use of IgM/IgG ratios in the clinics as a sign that an infection is recent (IgM only), ongoing acute (IgM and IgG) or occurred some time ago (IgG only), such interpretation of the serological data is an oversimplification of humoral immunity to infections, as opposed to model antigens. B cell plasmablasts develop rapidly following an acute infection and these responses provide much of the earliest measurable wave of serum antibodies. In contrast to the later-developing plasma cells developing from germinal center responses, which are mostly Ig-class-switched, these “extrafollicular” B cell responses produce IgM as well as class-switched IgG and IgA, depending on the involved lymph tissue and the cytokine milieu induced by the infection (Figure 1). Respiratory tract infections induce IgM, IgG and IgA, as indeed found for patients infected with SARS-CoV-2 (15, 31, 32). Based on current understanding of these mostly T-dependent responses, there is no expectation that IgM responses necessarily precedes class-switched responses, as class-switching is rapidly induced during the process of B cell activation and clonal expansion (3335), and thus prior to their differentiation to IgM or IgG-secreting cells. Given that extrafollicular B cell responses do not seem to require continued “T cell help” (36), production of IgM might be favored, as induction of AID, the enzyme facilitating class-switch recombination, is strongly induced via interaction through CD40-CD40L. What has been appreciated more recently is that B cell activation in the extrafollicular compartments of secondary lymph tissue can lead to the development of non-switched IgM memory B cells (37) and may even lead to long-lived IgM responses by plasma cells secreting IgM for extended periods of time (38, 39), again making a distinction between acute and later-stages of infection based on IgM/IgG ratios impossible.

Figure 1: Kinetics of primary and secondary antibody responses against acute viral infections.

Figure 1:

Early in the primary response, antibodies are contributed exclusively by extrafollicular plasmablasts and plasma cells (blue line). As the response progresses, germinal center-derived (green line) plasma cells with longer lifespans emerge and gradually increase in contribution to total antibody levels (purple line). Booster immunizations or re-infections trigger a memory B cell response, leading to a rapid rise in antibody levels through both germinal center-independent and -dependent mechanisms, and are often followed by a rapid decrease to levels just above pre-challenge concentrations.

In acute infections, such as a respiratory tract infections with influenza virus, IgM responses are induced and then rapidly lost, because IgM secreting LLPC are not induced. As the acute extrafollicular response resolves and the short-lived IgM and IgG or IgA-secreting plasmablasts die, germinal center-derived plasma cells replace these early-secreting cells, most if not all secreting class-switched antibodies, such as IgG and IgA. Since the half-life of IgM is only in the order of a few days (40), in contrast to that of IgG, which is in the order of 3–4 weeks depending on the isotypes or subclasses, serum IgM levels rapidly disappear. Thus, it is not surprising to see that IgM and IgG titers have been shown to rise together, or in some cases the IgG or IgA responses may even precede that of IgM in patients infected with SARS-COV-2 (15, 31, 32). The results suggest that, as expected, SARS-CoV-2 induces robust extrafollicular responses in the respiratory tract draining lymph tissue, driven in part by the local elaboration of inflammatory cytokines. Consistent with these conclusions, a recent study provides support for strong extrafollicular responses to SARS-CoV-2 (31). Their increased and possibly continued presence in severely ill patients underscores the nature of this humoral response as an “emergency supply system”, ensuring the rapid production of protective antibodies, both switched and non-switched. Asymptomatic patients and those with mild disease exhibit lower overall antibody responses, but do also possess neutralizing titers (4143). As convalescence begins, B cell responses shift from “emergency supply” to the development of long-lived responses in germinal center responses, which are geared towards developing protection from future re-encounters. We do not currently understand what drives the shift from the extrafollicular to the germinal center response, but based on preliminary data inflammatory signals are critical for the development of the EF response (Lam, J.H. & Baumgarth, N., in preparation).

Within the germinal center reaction, B cells undergo somatic hypermutation of their immunoglobulin genes and selection for those variants with higher affinity for antigen. High affinity variants are then fated for either memory B or plasma cell differentiation. Generally, as antibody affinity increases over time, so too does the lifespan and secretory capacity of plasma cells (4446). Consequently, relatively few plasma cells are required to confer protection against subsequent infections, provided antibodies are made against the key viral epitopes. As an example, passive transfer of a single microliter of West Nile virus immune serum, corresponding to just 3 antigen-specific long-lived plasma cells, is enough to protect a naïve mouse from an otherwise lethal inoculum (47). Plasma cell lifespan is variable based on the specific vaccine or infection, but as a general rule, the duration of antibody production is very long after natural exposures, as seen in responses to acute viruses such as mumps and measles (48). Long-lived plasma cells (LLPCs) are the sources of this durable antibody production (4954). Humoral immunity to chronic infections such as those caused by HIV and HPV, where viral antigens can persist in the host, are more complex (55). Given the acute nature of SARS-CoV-2 infections, chronic infections will not be discussed further here.

Humoral immunity in respiratory tract

Based on existing data on other acute respiratory tract infections, one would thus expect reductions or even disappearance of serum IgM and IgA responses to SARS-CoV-2 over time, while IgG titers should wane slower and remain elevated for extended periods of time (56). Unlike IgA, which is the dominant isotype in the upper respiratory tract, IgG is often considered to have minimal access to mucosal sites, yet this is not entirely accurate. First, although IgG cannot bind to the poly-Ig-receptor, which transcytoses dimeric IgA, FcRn can serve this function and promote IgG transport into lumenal secretions (57). As demonstrated by the highly successful intramuscular human papillomavirus vaccine, IgG can protect against mucosal infections through a combination of transcytosis and transudation (58). Second, serum IgA is usually monomeric rather than dimeric (59), and thus is unlikely to support lumenal defenses. Third, only the upper respiratory tract and large bronchi contain mucosa. The parenchyma of the lower respiratory tract lacks a mucosa, its barrier inhibiting oxygen exchange, and correspondingly, antiviral humoral responses in the lung are dominated by IgG rather than IgA (60). Thus, the rapid waning of IgA responses observed in COVID-19 patients (31, 61, 62), is not a priori a cause for concern in the background of stable neutralizing IgG titers. This IgG produced by LLPCs also have undergone extensive somatic hypermutation and affinity maturation, and can therefore provide strong protection against homologous virus challenges. The successful use of convalescent plasma from previous COVID-19 patients for the treatment of severely ill patients supports the conclusions that when patients have had robust infections with SARS-CoV-2, they will develop germinal center responses, and thus protective humoral immunity that can be expected to last for extended periods of time. Patients with only mild and localized infection in the upper respiratory tract may develop local tissue-resident antibody and/or cell-mediated immunity (7, 63), which would not be captured by serological testing. Critically ill individuals with ongoing dysregulated inflammatory responses may not shift from extrafollicular to germinal center responses and maybe unable to support germinal center responses.

Role of memory B cells in viral infections

Memory B cells tend to emerge earlier in the germinal center response, encode lower affinity antibodies than their long-lived plasma cell counterparts, and encompass a number of functionally distinct subsets (37, 6470). Immunoglobulin class-switched memory B cells are thought of as the first responders when pathogens evade pre-existing serum antibodies (37). This is likely because these cells can bypass the requirement for T cell help in response to viral infections (71). In mice, these memory B cells can be further distinguished by expression of the markers CD80 and PD-L2 (37, 64, 66, 67). Importantly, similar to responses seen during a primary infection, these memory B cells generate the transiently-induced extrafollicular “emergency” antibody responses, likely driven by the same inflammatory signals that initiated this responses during a primary infection, but now derived from a previously selected and clonally-expanded population of B cells. Recall responses may also be driven in part by transcriptional programs that bias towards transience more so than primary responses (7274). The distinction between primary and memory B cell responses becomes particularly important in the context of prior coronavirus studies.

Duration of humoral immunity after coronavirus and other acute viral infections

Duration of immunity to common coronaviruses

Infectious coronaviruses with human tropism can be stratified into alpha and beta lineages. Whereas the most pathogenic coronaviruses, including MERS, SARS-CoV-1, and SARS-CoV-2 fall within the latter, so too do less dangerous and more common strains, including OC43 and HKU1 (75). The most common alpha coronaviruses include 229E and NL63, both of which cause a relatively mild “cold”. Because of the mildness of symptoms caused by the common human coronaviruses, much of our information has come from early challenge studies of human volunteers using live virus, dating back to the 1960s and before modern tools to distinguish genotypes were available (76).

The seroprevalence for the common human coronaviruses is well over 90% (77). Thus, it is not feasible to identify immunologically naïve adults for challenge studies. Adding further complications, there is substantial serological cross-reactivity across different coronaviruses within and even across alpha and beta lineages (78). Thus, challenge studies in these settings almost certainly measure memory rather than primary responses, without a means to determine whether the challenges are homologous or heterologous. In early studies, challenge of volunteers with the 229E coronavirus strain led to complete resistance to re-infection and shedding by the same virus for at least one year (79). Of note, despite substantial serological cross-reactivity, this same level of resistance was not observed when subjects were re-challenged with heterologous alpha coronaviruses with similarity to 229E (79). These data emphasize the important distinction between viral serotypes and genotypes. Durable immunity to homologous strains is almost certain; immunity to heterologous strains is less certain.

A widely cited study followed these early experiments in which antibody titers were followed after experimental challenge of human volunteers, demonstrating that antibody production returns to near pre-challenge levels after only 1 year (80). This observation has widely been interpreted as evidence that immunity to coronaviruses is intrinsically transient. Yet, this interpretation should be tempered for several reasons. First, upon a second challenge one year after the first, none of the subjects developed symptomatic disease and viral shedding was transient and minimal. Second, the pre-existing baseline titers against 229E were high for all experimental groups prior to the challenge. In the productively infected subjects, antibody kinetics rose and fell quickly before returning to slightly above this baseline, consistent with the expectation of memory B cell responses. Importantly, the group of subjects that could not be productively infected maintained very high antibody titers that did not change before or after attempted infection, suggesting very durable protective humoral immunity. Finally, as with all challenge experiments, the experimental inoculum was likely higher than that which occurs in natural exposures. Together, these findings imply that immunity to homologous common coronaviruses lasts for longer than the single year that is often stated in review articles and in the media.

Why, then, do we keep getting infected with common coronaviruses so often (81, 82)? The answer may lie in the underappreciated genetic diversity of coronavirus strains (8385). The mutation rate for both SARS-CoV2 and common coronaviruses is relatively low (for NL63, the estimated substitution rate is 3 × 10−4 per site per year in the Spike protein), but some of the common coronaviruses have been circulating amongst the human population for an estimated 1000 years (83, 86). Given the near universal seroprevalence for common coronaviruses, presumably there exists intense selective pressure to evade these defenses. Indeed, early studies suggested that coronaviruses of the same serogroup may be able to escape heterologous immunity (79). The extent to which prior coronavirus infections impact heterologous immunity to SARS-CoV-2 remains under investigation (22, 25, 87).

Duration of immunity to SARS-CoV-1 and SARS-CoV-2

Intentional challenge studies are of course not ethical for deadly strains such as MERS and SARS-CoV-1. Thus, surrogates must be relied upon to estimate the duration of immunity. Studies on SARS-CoV-1 demonstrated that virus-specific IgG antibody titers peaked at month 4 and their geometric mean reciprocal titers declined 7-fold and 10-fold at months 24 and 36, respectively (88). Based on these data, it was concluded that antibody production lasts for only 1–3 years after clearance of infection. These estimates, however, are problematic when extrapolated from the early phases of the response when many short-lived plasma cells are dying, which as discussed provide the early wave of antibodies. For most acute challenges, a relatively stable nadir is eventually achieved of antibodies continuously produced by long-lived plasma cells (49). Thus, the decay of antibody production during the course of a response is not linear because it is contributed by distinct plasma cell subsets (both short-lived and long-lived), and the duration of immunity can therefore only be calculated over longer periods of time (Figure 1). Indeed, recent studies reported that SARS-CoV-1-neutralizing antibodies could still be detected 12–17 years after the infection was resolved (89, 90). Interestingly, antibody titers were low as measured by standard ELISAs, demonstrating that very small quantities of high-affinity antibodies can prevent infection, at least in vitro. Because SARS-CoV-1 infected relatively few people and no longer circulates, there is no way to epidemiologically determine if people resist re-infection so long into convalescence.

The SARS-CoV-2 pandemic began only a few months ago. Thus, by definition, humoral responses in those infected are still in the early phases. Several SARS-CoV-2 studies have suggested rapid waning of immunity and antibody responses against the nucleocapsid protein, especially in those who recovered from mildly symptomatic disease (41, 91). For the same reasons as discussed above for SARS-CoV-1, such conclusions are premature in our view. All antibody responses, even exceptionally durable ones such as against measles, show an initial decline not inconsistent with what has been observed for SARS-CoV-2 thus far (48). Moreover, neutralizing antibodies to SARS-CoV-2, which presumably bind the spike protein, remained very high in the same study (41). The discrepancy might reflect differences in either assay sensitivity or antibody responses to nucleocapsid vs. spike protein. Based on the fundamental principles of B cell response regulation that we outlined above, we consider it very unlikely that immunity to SARS-CoV-2 would be lost already. At present, serum antibody titers are clearly important, but a comprehensive assessment of protective SARS-CoV-2 immunity will be more informative. It is likely to include both humoral and cellular measurements that will include mucosal and serum antibodies as well as cellular measures that include circulating and tissue resident memory T and B cells.

Durability of antibody responses to other acute respiratory viruses

Aside from coronaviruses, a wealth of knowledge has also been gained for other acute respiratory infections. Respiratory syncytial virus (RSV) is often cited as an example of a natural infection conferring only short-term immunity. Yet, a review of the literature suggests more complexity than is appreciated. After infection, neutralizing antibodies do indeed decline initially (92), as is seen in all humoral responses irrespective of the ultimate duration of immunity (48). Yet when followed over the course of several years, protection against subsequent infection is the norm and later timepoints suggest a relatively stable nadir of antibodies in most individuals, the levels of which correlate with the extent of protection (93, 94). For example, higher pre-existing serum or nasal antibodies precluded RSV infections in the challenge studies (93, 95). Thus, immunity to RSV is the norm in the general adult population, since severe RSV infections are uncommon until advanced age (96, 97). Immunological considerations in the elderly are an important concern that we will discuss more in the next section. Thus, though it is clear that lifelong and complete protection against RSV from a single exposure is unlikely, the focus on only those individuals who rapidly lose protective antibody titers provides an incomplete picture of the typical response. As another example of an acute respiratory infection, studies of adults nearly 90 years after the 1918 influenza virus pandemic demonstrated high titers of serum neutralizing antibodies to the homologous strain (98). The remarkably durable immunity in individuals likely exposed as children probably arose from the primary response. At least for other vaccines, repeated stimulation of memory B cells is not required for serum antibody maintenance (52), demonstrating that primary infections and immunizations can confer durable antibody production. In further support of this interpretation, young children with no pre-existing immunity immunized with an MF59-adjuvanted influenza vaccine demonstrated robust and durable immunity (99). As a final example, homologous rhinovirus challenge studies demonstrated immunity for at least a year (100). Together, these data collectively demonstrate that durable protection against homotypic challenges following acute viral infections is the norm. Following natural infections with pathogens such as influenza viruses and rhinoviruses, the major factor imparting a loss of immunity is not short-lived antibody production, but rather the emergence of viral serotypes and genotypes that evade these defenses. Thus far, the rate of emergence of new genetic variants of SARS-CoV-2 has been slow (101), yet it is certainly possible that after widespread vaccination efforts and thus increased immune selective pressure, this rate will increase.

Immune responses in vulnerable populations against SARS-CoV-2 and against the potential COVID-19 vaccines

Though immunity following natural SARS-CoV-2 infection is likely to be durable, this is less clear for immunity to vaccines, especially given the experimental nature of many of these platforms (48, 102). These considerations are especially important when considering populations at special risk of severe disease and death from COVID-19. This includes those over 60 (103), as well as persons with underlying medical conditions (diabetes, hypertension, chronic pulmonary diseases etc.), although by now it is very clear that SARS-CoV-2 can and will strike outside of these categories and that it can be fatal for young people with no underlying conditions at all. That fact highlights the limits of our knowledge and understanding of COVID-19 pathogenesis, an issue that would greatly benefit from a reliable animal model that recapitulates the main clinical findings from humans.

This issue notwithstanding, the questions on the immune responses in vulnerable populations, the durability of immunity in these populations and their protection by vaccination all remain at the forefront of both research and clinical medicine. Due to the space constraints, and due to the current state of knowledge, we will discuss these issues from the perspective of older adults, at the moment not considering the underlying conditions. It is worth noting that pronounced lymphopenia follows severe COVID-19 (104). Whether that reflects a lymphotoxic effect of SARS-CoV-2 or the SARS-CoV-2-modulated environment, or (more likely) redistribution of lymphocytes to target organs, such as the lung, or some other mechanism, remains to be seen. Usually, lymphopenia would bode ill for the immune responses in older adults, because older adults already lose substantial numbers of naïve CD8, and to a lesser extent CD4 and B cells (105, 106). Indeed, primary adaptive immune responses to new infections (T as well as B) in older adults are reduced, delayed and suboptimally coordinated (106). Interestingly, the literature so far has not supported such a common scenario in the case of SARS-CoV-2. Antibody responses in older adults were quantitatively robust and at least equivalent, if not superior to, those of their younger counterparts (15, 107), While the quality of these responses also appears comparable, this remains to be examined in greater detail with regard to affinity/avidity, repertoire breadth and functional capacity. Instead, it appears that the severity of infection seems to dictate the extent and intensity of the immune response (15, 107), which, by itself, does not correlate to the disease outcome. At the present, it appears that the coordination of the innate immune response may be key to successful containment of SARS-CoV-2 (108), and we have no information on whether or not this coordination may be adversely affected by the age of the subject.

Importantly, robust responses to SARS-CoV-2 in older adults give us hope that this population can be protected by both immunomodulation and vaccination. Vaccination in the elderly usually suffers from the same problems of other primary immune responses in the elderly, and is known to be efficacious in only half or even fewer of the individuals (17–51%, depending on the season), in the case of seasonal influenza vaccine, as compared to 70–85% in younger populations (109). However, new generation of vaccines, adjuvanted with patter recognition receptor (PRR) agonists, have been shown to induce highly efficient immune responses in older adults, with the varicella-zoster virus vaccine Shingrix®, conferring strong antibody immunity to >97% adults over 65 years of age (110). We therefore hold high hope that vaccine manufacturers will keep this in mind when designing SARS-CoV-2 vaccines, to protect broad swaths of vulnerable populations across the world.

Conclusions

The purpose of this article is not to paint an excessively rosy picture of the SARS-CoV-2/COVID-19 pandemic. Rather, the intent is to offer an objective perspective of antibody immunity. Doing so allows for a proper calibration of likelihoods, expectations, and uncertainty, thereby balancing particularly worrisome aspects of the virus to those that are of much lesser concern. There are many highly problematic immunopathological and epidemiological aspects of this virus, but we do not believe that subversion of fundamental immunological principles is among them.

Acknowledgments

Funding Sources

Funding was provided by the National Institutes of Health: R01AI099108 (D.B.), R01AI121252 (F.E.L.), R01AI117890 (N.B.) R01AI148652 (N.B.) and R37AG020719 (J.N.Z.).

Conflicts of interest

D.B. has intellectual property licensed by Sana Biotechnology. F.E.L. is a founder of MicroB-plex, Inc. and receives research funding from Genentech. J.N.Z. is on the scientific advisory board of and receives research funding from Young Blood Inc.

References

  • 1.Zhou P, Yang X-L, Wang X-G, Hu B, Zhang L, Zhang W, Si H-R, Zhu Y, Li B, Huang C-L, Chen H-D, Chen J, Luo Y, Guo H, Jiang R-D, Liu M-Q, Chen Y, Shen X-R, Wang X, Zheng X-S, Zhao K, Chen Q-J, Deng F, Liu L-L, Yan B, Zhan F-X, Wang Y-Y, Xiao G-F, and Shi Z-L 2020. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 579: 270–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Wu F, Zhao S, Yu B, Chen Y-M, Wang W, Song Z-G, Hu Y, Tao Z-W, Tian J-H, Pei Y-Y, Yuan M-L, Zhang Y-L, Dai F-H, Liu Y, Wang Q-M, Zheng J-J, Xu L, Holmes EC, and Zhang Y-Z 2020. A new coronavirus associated with human respiratory disease in China. Nature 579: 265–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.World Health Organization. Advice on the use of masks in the context of COVID-19: interim guidance, 6 April 2020.
  • 4.World Health Organization. 2020. “Immunity passports” in the context of COVID-19.
  • 5.Centers for Disease Control and Prevention. 2020. Interim Considerations for Institutions of Higher Education Administrators for SARS-CoV-2 Testing. Centers for Disease Control and Prevention. [Google Scholar]
  • 6.Masopust D, Vezys V, Marzo AL, and Lefrançois L 2001. Preferential Localization of Effector Memory Cells in Nonlymphoid Tissue. Science 291: 2413–2417. [DOI] [PubMed] [Google Scholar]
  • 7.Allie SR, Bradley JE, Mudunuru U, Schultz MD, Graf BA, Lund FE, and Randall TD 2019. The establishment of resident memory B cells in the lung requires local antigen encounter. Nature Immunology 20: 97–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cooper MA, Elliott JM, Keyel PA, Yang L, Carrero JA, and Yokoyama WM 2009. Cytokine-induced memory-like natural killer cells. PNAS 106: 1915–1919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Sun JC, Beilke JN, and Lanier LL 2009. Adaptive immune features of natural killer cells. Nature 457: 557–561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Netea MG, Quintin J, and van der Meer JWM 2011. Trained Immunity: A Memory for Innate Host Defense. Cell Host & Microbe 9: 355–361. [DOI] [PubMed] [Google Scholar]
  • 11.Korean Centers for Disease Control. 2020. Findings from investigation and analysis of re-positive cases (http://www.cdc.go.kr).
  • 12.Xiao AT, Tong YX, and Zhang S 2020. False negative of RT-PCR and prolonged nucleic acid conversion in COVID-19: Rather than recurrence. J. Med. Virol. epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Yang C, Jiang M, Wang X, Tang X, Fang S, Li H, Zuo L, Jiang Y, Zhong Y, Chen Q, Zheng C, Wang L, Wu S, Wu W, Liu H, Yuan J, Liao X, Zhang Z, Lin Y, Geng Y, Zhang H, Zheng H, Wan M, Lu L, Ren X, Cui Y, Zou X, Feng T, Xia J, Yang R, Liu Y, Mei S, Li B, Yang Z, and Hu Q 2020. Viral RNA level, serum antibody responses, and transmission risk in discharged COVID-19 patients with recurrent positive SARS-CoV-2 RNA test results: a population-based observational cohort study. medRxiv 2020.07.21.20125138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zinkernagel RM, and Hengartner H 2006. Protective ‘immunity’ by pre-existent neutralizing antibody titers and preactivated T cells but not by so-called ‘immunological memory.’ Immunological Reviews 211: 310–319. [DOI] [PubMed] [Google Scholar]
  • 15.Long Q-X, Liu B-Z, Deng H-J, Wu G-C, Deng K, Chen Y-K, Liao P, Qiu J-F, Lin Y, Cai X-F, Wang D-Q, Hu Y, Ren J-H, Tang N, Xu Y-Y, Yu L-H, Mo Z, Gong F, Zhang X-L, Tian W-G, Hu L, Zhang X-X, Xiang J-L, Du H-X, Liu H-W, Lang C-H, Luo X-H, Wu S-B, Cui X-P, Zhou Z, Zhu M-M, Wang J, Xue C-J, Li X-F, Wang L, Li Z-J, Wang K, Niu C-C, Yang Q-J, Tang X-J, Zhang Y, Liu X-M, Li J-J, Zhang D-C, Zhang F, Liu P, Yuan J, Li Q, Hu J-L, Chen J, and Huang A-L 2020. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat Med 26: 845–848. [DOI] [PubMed] [Google Scholar]
  • 16.Duan K, Liu B, Li C, Zhang H, Yu T, Qu J, Zhou M, Chen L, Meng S, Hu Y, Peng C, Yuan M, Huang J, Wang Z, Yu J, Gao X, Wang D, Yu X, Li L, Zhang J, Wu X, Li B, Xu Y, Chen W, Peng Y, Hu Y, Lin L, Liu X, Huang S, Zhou Z, Zhang L, Wang Y, Zhang Z, Deng K, Xia Z, Gong Q, Zhang W, Zheng X, Liu Y, Yang H, Zhou D, Yu D, Hou J, Shi Z, Chen S, Chen Z, Zhang X, and Yang X 2020. Effectiveness of convalescent plasma therapy in severe COVID-19 patients. Proc Natl Acad Sci USA 117: 9490–9496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Shen C, Wang Z, Zhao F, Yang Y, Li J, Yuan J, Wang F, Li D, Yang M, Xing L, Wei J, Xiao H, Yang Y, Qu J, Qing L, Chen L, Xu Z, Peng L, Li Y, Zheng H, Chen F, Huang K, Jiang Y, Liu D, Zhang Z, Liu Y, and Liu L 2020. Treatment of 5 Critically Ill Patients With COVID-19 With Convalescent Plasma. JAMA 323: 1582–1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Liu STH, Lin H-M, Baine I, Wajnberg A, Gumprecht JP, Rahman F, Rodriguez D, Tandon P, Bassily-Marcus A, Bander J, Sanky C, Dupper A, Zheng A, Altman DR, Chen BK, Krammer F, Mendu DR, Firpo-Betancourt A, Levin MA, Bagiella E, Casadevall A, Cordon-Cardo C, Jhang JS, Arinsburg SA, Reich DL, Aberg JA, and Bouvier NM 2020. Convalescent plasma treatment of severe COVID-19: A matched control study. medRxiv 2020.05.20.20102236. [DOI] [PubMed] [Google Scholar]
  • 19.Li L, Zhang W, Hu Y, Tong X, Zheng S, Yang J, Kong Y, Ren L, Wei Q, Mei H, Hu C, Tao C, Yang R, Wang J, Yu Y, Guo Y, Wu X, Xu Z, Zeng L, Xiong N, Chen L, Wang J, Man N, Liu Y, Xu H, Deng E, Zhang X, Li C, Wang C, Su S, Zhang L, Wang J, Wu Y, and Liu Z 2020. Effect of Convalescent Plasma Therapy on Time to Clinical Improvement in Patients With Severe and Life-threatening COVID-19: A Randomized Clinical Trial. JAMA online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Chandrashekar A, Liu J, Martinot AJ, McMahan K, Mercado NB, Peter L, Tostanoski LH, Yu J, Maliga Z, Nekorchuk M, Busman-Sahay K, Terry M, Wrijil LM, Ducat S, Martinez DR, Atyeo C, Fischinger S, Burke JS, Slein MD, Pessaint L, Ry AV, Greenhouse J, Taylor T, Blade K, Cook A, Finneyfrock B, Brown R, Teow E, Velasco J, Zahn R, Wegmann F, Abbink P, Bondzie EA, Dagotto G, Gebre MS, He X, Jacob-Dolan C, Kordana N, Li Z, Lifton MA, Mahrokhian SH, Maxfield LF, Nityanandam R, Nkolola JP, Schmidt AG, Miller AD, Baric RS, Alter G, Sorger PK, Estes JD, Andersen H, Lewis MG, and Barouch DH 2020. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bao L, Deng W, Gao H, Xiao C, Liu J, Xue J, Lv Q, Liu J, Yu P, Xu Y, Qi F, Qu Y, Li F, Xiang Z, Yu H, Gong S, Liu M, Wang G, Wang S, Song Z, Zhao W, Han Y, Zhao L, Liu X, Wei Q, and Qin C 2020. Reinfection could not occur in SARS-CoV-2 infected rhesus macaques. bioRxiv 2020.03.13.990226. [Google Scholar]
  • 22.Sekine T, Perez-Potti A, Rivera-Ballesteros O, Strålin K, Gorin J-B, Olsson A, Llewellyn-Lacey S, Kamal H, Bogdanovic G, Muschiol S, Wullimann DJ, Kammann T, Emgård J, Parrot T, Folkesson E, Rooyackers O, Eriksson LI, Sönnerborg A, Allander T, Albert J, Nielsen M, Klingström J, Gredmark-Russ S, Björkström NK, Sandberg JK, Price DA, Ljunggren H-G, Aleman S, Buggert M, and K. C.−19 S. Group. 2020. Robust T cell immunity in convalescent individuals with asymptomatic or mild COVID-19. bioRxiv 2020.06.29.174888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Pollán M, Pérez-Gómez B, Pastor-Barriuso R, Oteo J, Hernán MA, Pérez-Olmeda M, Sanmartín JL, Fernández-García A, Cruz I, de Larrea NF, Molina M, Rodríguez-Cabrera F, Martín M, Merino-Amador P, Paniagua JL, Muñoz-Montalvo JF, Blanco F, Yotti R, Blanco F, Fernández RG, Martín M, Navarro SM, Molina M, Muñoz-Montalvo JF, Hernández MS, Sanmartín JL, Cuenca-Estrella M, Yotti R, Paniagua JL, de Larrea NF, Fernández-Navarro P, Pastor-Barriuso R, Pérez-Gómez B, Pollán M, Avellón A, Fedele G, Fernández-García A, Iglesias JO, Olmeda MTP, Cruz I, Martinez MEF, Rodríguez-Cabrera FD, Hernán MA, Fernández SP, Aguirre JMR, Marí JMN, Borrás BP, Jiménez ABP, Rodríguez-Iglesias M, Gascón AMC, Alcaine MLL, Suárez ID, Álvarez OS, Pérez MR, Sanchís MC, Gomila CJV, Saladrigas LC, Fernández AH, Oliver A, Feliciano EC, Quintana MNG, Fernández JMB, Betancor MAH, Febles MH, Martín LM, López L-ML, Miota TU, Población IDB, Pérez MSC, Fernández MNV, Enríquez TM, Arranz MV, González MD-G, Fernández-Natal I, Lobón GM, Bellido JLM, Ciruela P, i Casals AM, Botías MD, Maeso MAM, del Campo DP, de Castro AF, Ramírez RL, Retamosa MFE, González MR, Lobeiras MSB, Losada AF, Aguilera A, Bou G, Caro Y, Marauri N, Blanco LMS, González I. del C, Pascual MH, Fernández RA, Merino-Amador P, Castro NC, Lizcano AT, Almagro CR, Hernández MS, Elizaga NA, Sanz ME, Baquedano CE, Bascaran AB, Tamayo SI, Otazua LE, Benarroch RB, Flores JL, and de la Villa AV 2020. Prevalence of SARS-CoV-2 in Spain (ENE-COVID): a nationwide, population-based seroepidemiological study. The Lancet online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Liu T, Wu S, Tao H, Zeng G, Zhou F, Guo F, and Wang X 2020. Prevalence of IgG antibodies to SARS-CoV-2 in Wuhan - implications for the ability to produce long-lasting protective antibodies against SARS-CoV-2. medRxiv 2020.06.13.20130252. [Google Scholar]
  • 25.Grifoni A, Weiskopf D, Ramirez SI, Mateus J, Dan JM, Moderbacher CR, Rawlings SA, Sutherland A, Premkumar L, Jadi RS, Marrama D, de Silva AM, Frazier A, Carlin AF, Greenbaum JA, Peters B, Krammer F, Smith DM, Crotty S, and Sette A 2020. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 181: 1489–1501.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Premkumar L, Segovia-Chumbez B, Jadi R, Martinez DR, Raut R, Markmann AJ, Cornaby C, Bartelt L, Weiss S, Park Y, Edwards CE, Weimer E, Scherer EM, Rouphael N, Edupuganti S, Weiskopf D, Tse LV, Hou YJ, Margolis D, Sette A, Collins MH, Schmitz J, Baric RS, and de Silva AM 2020. The receptor-binding domain of the viral spike protein is an immunodominant and highly specific target of antibodies in SARS-CoV-2 patients. Science Immunology 5: eabc8413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Amanat F, Stadlbauer D, Strohmeier S, Nguyen THO, Chromikova V, McMahon M, Jiang K, Arunkumar GA, Jurczyszak D, Polanco J, Bermudez-Gonzalez M, Kleiner G, Aydillo T, Miorin L, Fierer DS, Lugo LA, Kojic EM, Stoever J, Liu STH, Cunningham-Rundles C, Felgner PL, Moran T, García-Sastre A, Caplivski D, Cheng AC, Kedzierska K, Vapalahti O, Hepojoki JM, Simon V, and Krammer F 2020. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nature Medicine 1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Meyer B, Torriani G, Yerly S, Mazza L, Calame A, Arm-Vernez I, Zimmer G, Agoritsas T, Stirnemann J, Spechbach H, Guessous I, Stringhini S, Pugin J, Roux-Lombard P, Fontao L, Siegrist C-A, Eckerle I, Vuilleumier N, and Kaiser L 2020. Validation of a commercially available SARS-CoV-2 serological immunoassay. Clinical Microbiology and Infection online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Testing the Tests: COVID-19 Antibody Assays Scrutinized for Accuracy by UCSF, UC Berkeley Researchers. Testing the Tests: COVID-19 Antibody Assays Scrutinized for Accuracy by UCSF, UC Berkeley Researchers | UC San Francisco. [Google Scholar]
  • 30.Bryan A, Pepper G, Wener MH, Fink SL, Morishima C, Chaudhary A, Jerome KR, Mathias PC, and Greninger AL 2020. Performance Characteristics of the Abbott Architect SARS-CoV-2 IgG Assay and Seroprevalence in Boise, Idaho. Journal of Clinical Microbiology online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Woodruff M, Ramonell R, Cashman K, Nguyen D, Saini A, Haddad N, Ley A, Kyu S, Howell JC, Ozturk T, Lee S, Chen W, Estrada J, Morrison-Porter A, Derrico A, Anam F, Sharma M, Wu H, Le S, Jenks S, Tipton CM, Hu W, Lee FE-H, and Sanz I 2020. Dominant extrafollicular B cell responses in severe COVID-19 disease correlate with robust viral-specific antibody production but poor clinical outcomes. medRxiv 2020.04.29.20083717. [Google Scholar]
  • 32.Guo L, Ren L, Yang S, Xiao M, Chang D, Yang F, Dela Cruz CS, Wang Y, Wu C, Xiao Y, Zhang L, Han L, Dang S, Xu Y, Yang Q-W, Xu S-Y, Zhu H-D, Xu Y-C, Jin Q, Sharma L, Wang L, and Wang J Profiling Early Humoral Response to Diagnose Novel Coronavirus Disease (COVID-19). Clin Infect Dis 71: 778–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Marshall JL, Zhang Y, Pallan L, et al. Early B blasts acquire a capacity for Ig class switch recombination that is lost as they become plasmablasts. Eur J Immunol. 2011;41(12):3506–3512. doi: 10.1002/eji.201141762. [DOI] [PubMed] [Google Scholar]
  • 34.MacLennan IC, Toellner KM, Cunningham AF, et al. Extrafollicular antibody responses. Immunol Rev. 2003;194:8–18. doi: 10.1034/j.1600-065x.2003.00058.x. [DOI] [PubMed] [Google Scholar]
  • 35.Roco JA, Mesin L, Binder SC, Nefzger C, Gonzalez-Figueroa P, Canete PF, Ellyard J, Shen Q, Robert PA, Cappello J, Vohra H, Zhang Y, Nowosad CR, Schiepers A, Corcoran LM, Toellner K-M, Polo JM, Meyer-Hermann M, Victora GD, and Vinuesa CG 2019. Class-Switch Recombination Occurs Infrequently in Germinal Centers. Immunity 51: 337–350.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.MacLennan IC, Toellner KM, Cunningham AF, et al. Extrafollicular antibody responses. Immunol Rev. 2003;194:8–18. doi: 10.1034/j.1600-065x.2003.00058.x.. [DOI] [PubMed] [Google Scholar]
  • 37.Pape KA, Taylor JJ, Maul RW, Gearhart PJ, and Jenkins MK 2011. Different B Cell Populations Mediate Early and Late Memory During an Endogenous Immune Response. Science 331: 1203–1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Racine R, McLaughlin M, Jones DD, et al. IgM production by bone marrow plasmablasts contributes to long-term protection against intracellular bacterial infection. J Immunol. 2011;186(2):1011–1021. doi: 10.4049/jimmunol.1002836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bohannon C, Powers R, Satyabhama L, et al. Long-lived antigen-induced IgM plasma cells demonstrate somatic mutations and contribute to long-term protection [published correction appears in Nat Commun. 2016 Aug 16;7:12687. Lee, Francis Eun-Hyung [Lee, Frances Eun-Hyung]]. Nat Commun. 2016;7:11826 Published 2016 Jun 7. doi: 10.1038/ncomms11826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Brekke OH, Sandlie I. Therapeutic antibodies for human diseases at the dawn of the twenty-first century [published correction appears in Nat Rev Drug Discov. 2003 Mar;2(3):240.]. Nat Rev Drug Discov 2003;2(1):52–62. doi: 10.1038/nrd984. [DOI] [PubMed] [Google Scholar]
  • 41.Long Q-X, Tang X-J, Shi Q-L, Li Q, Deng H-J, Yuan J, Hu J-L, Xu W, Zhang Y, Lv F-J, Su K, Zhang F, Gong J, Wu B, Liu X-M, Li J-J, Qiu J-F, Chen J, and Huang A-L 2020. Clinical and immunological assessment of asymptomatic SARS-CoV-2 infections. Nat. Med. online ahead of print. [DOI] [PubMed] [Google Scholar]
  • 42.Ko J-H, Joo E-J, Park S-J, Baek JY, Kim WD, Jee J, Kim CJ, Jeong C, Kim Y-J, Shon HJ, Kang E-S, Choi YK, and Peck KR 2020. Neutralizing Antibody Production in Asymptomatic and Mild COVID-19 Patients, in Comparison with Pneumonic COVID-19 Patients. Journal of Clinical Medicine 9: 2268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Choe PG, Kang CK, Suh HJ, Jung J, Kang E, Lee SY, Song K-H, Kim HB, Kim NJ, Park WB, Kim ES, and Oh M Early Release - Antibody Responses to SARS-CoV-2 at 8 Weeks Postinfection in Asymptomatic Patients - Volume 26, Number 10—October 2020. - Emerging Infectious Diseases journal - CDC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Weisel FJ, Zuccarino-Catania GV, Chikina M, and Shlomchik MJ 2016. A Temporal Switch in the Germinal Center Determines Differential Output of Memory B and Plasma Cells. Immunity 44: 116–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lam WY, Jash A, Yao C-H, D’Souza L, Wong R, Nunley RM, Meares GP, Patti GJ, and Bhattacharya D 2018. Metabolic and Transcriptional Modules Independently Diversify Plasma Cell Lifespan and Function. Cell Reports 24: 2479–2492.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lam WY, Becker AM, Kennerly KM, Wong R, Curtis JD, Llufrio EM, McCommis KS, Fahrmann J, Pizzato HA, Nunley RM, Lee J, Wolfgang MJ, Patti GJ, Finck BN, Pearce EL, and Bhattacharya D 2016. Mitochondrial Pyruvate Import Promotes Long-Term Survival of Antibody-Secreting Plasma Cells. Immunity 45: 60–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Purtha WE, Tedder TF, Johnson S, Bhattacharya D, and Diamond MS 2011. Memory B cells, but not long-lived plasma cells, possess antigen specificities for viral escape mutants. J Exp Med 208: 2599–2606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Amanna IJ 2007. Duration of Humoral Immunity to Common Viral and Vaccine Antigens. The New England Journal of Medicine 357: 1903–1915. [DOI] [PubMed] [Google Scholar]
  • 49.Halliley JL, Tipton CM, Liesveld J, Rosenberg AF, Darce J, Gregoretti IV, Popova L, Kaminiski D, Fucile CF, Albizua I, Kyu S, Chiang K-Y, Bradley KT, Burack R, Slifka M, Hammarlund E, Wu H, Zhao L, Walsh EE, Falsey AR, Randall TD, Cheung WC, Sanz I, and Lee FE-H 2015. Long-Lived Plasma Cells Are Contained within the CD19−CD38hiCD138+ Subset in Human Bone Marrow. Immunity 43: 132–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mei HE, Wirries I, Frölich D, Brisslert M, Giesecke C, Grün JR, Alexander T, Schmidt S, Luda K, Kühl AA, Engelmann R, Dürr M, Scheel T, Bokarewa M, Perka C, Radbruch A, and Dörner T 2015. A unique population of IgG-expressing plasma cells lacking CD19 is enriched in human bone marrow. Blood 125: 1739–1748. [DOI] [PubMed] [Google Scholar]
  • 51.Landsverk OJB, Snir O, Casado RB, Richter L, Mold JE, Réu P, Horneland R, Paulsen V, Yaqub S, Aandahl EM, Øyen OM, Thorarensen HS, Salehpour M, Possnert G, Frisén J, Sollid LM, Baekkevold ES, and Jahnsen FL 2017. Antibody-secreting plasma cells persist for decades in human intestine. J Exp Med 214: 309–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Cambridge G, Leandro MJ, Edwards JCW, Ehrenstein MR, Salden M, Bodman‐Smith M, and Webster ADB 2003. Serologic changes following B lymphocyte depletion therapy for rheumatoid arthritis. Arthritis & Rheumatism 48: 2146–2154. [DOI] [PubMed] [Google Scholar]
  • 53.Slifka MK, Antia R, Whitmire JK, and Ahmed R 1998. Humoral Immunity Due to Long-Lived Plasma Cells. Immunity 8: 363–372. [DOI] [PubMed] [Google Scholar]
  • 54.Manz RA, Thiel A, and Radbruch A 1997. Lifetime of plasma cells in the bone marrow. Nature 388: 133–134. [DOI] [PubMed] [Google Scholar]
  • 55.Wong R, and Bhattacharya D 2019. Basics of memory B-cell responses: lessons from and for the real world. Immunology 156: 120–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Borremans B, Gamble A, Prager KC, Helman SK, McClain AM, Cox C, Savage V, and Lloyd-Smith JO 2020. Quantifying antibody kinetics and RNA shedding during early-phase SARS-CoV-2 infection. medRxiv 2020.05.15.20103275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yoshida M, Claypool SM, Wagner JS, Mizoguchi E, Mizoguchi A, Roopenian DC, Lencer WI, and Blumberg RS 2004. Human Neonatal Fc Receptor Mediates Transport of IgG into Luminal Secretions for Delivery of Antigens to Mucosal Dendritic Cells. Immunity 20: 769–783. [DOI] [PubMed] [Google Scholar]
  • 58.2007. Quadrivalent Vaccine against Human Papillomavirus to Prevent High-Grade Cervical Lesions. New England Journal of Medicine 356: 1915–1927. [DOI] [PubMed] [Google Scholar]
  • 59.Mestecky J 1988. Immunobiology of IgA. Am. J. Kidney Dis. 12: 378–383. [DOI] [PubMed] [Google Scholar]
  • 60.Jones PD, and Ada GL 1987. Persistence of influenza virus-specific antibody-secreting cells and B-cell memory after primary murine influenza virus infection. Cell. Immunol. 109: 53–64. [DOI] [PubMed] [Google Scholar]
  • 61.Iyer AS, Jones FK, Nodoushania A, Kelly M, Becker M, Slater D, Mills R, Teng E, Kamruzzaman M, Garcia-Beltran WF, Astudillo M, Yang D, Miller TE, Oiver E, Fischinger S, Atyeo C, Iafrate AJ, Calderwood SB, Lauer SA, Yu J, Li Z, Feldman J, Hauser BM, Cardonna TM, Branda JA, Turbett SE, LaRocque RC, Mellon G, Barouch DH, Schmidt AG, Azman AS, Alter G, Ryan ET, Harris JB, and Charles RC 2020. Dynamics and significance of the antibody response to SARS-CoV-2 infection. medRxiv 2020.07.18.20155374. [Google Scholar]
  • 62.Cervia C, Nilsson J, Zurbuchen Y, Valaperti A, Schreiner J, Wolfensberger A, Raeber ME, Adamo S, Emmenegger M, Hasler S, Bosshard PP, Cecco ED, Bächli E, Rudiger A, Stüssi-Helbling M, Huber LC, Zinkernagel AS, Schaer DJ, Aguzzi A, Held U, Probst-Müller E, Rampini SK, and Boyman O 2020. Systemic and mucosal antibody secretion specific to SARS-CoV-2 during mild versus severe COVID-19. bioRxiv 2020.05.21.108308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Van Braeckel-Budimir N, Harty JT. Influenza-induced lung Trm: not all memories last forever. Immunol Cell Biol. 2017;95(8):651–655. doi: 10.1038/icb.2017.32. [DOI] [PubMed] [Google Scholar]
  • 64.Zuccarino-Catania GV, Sadanand S, Weisel FJ, Tomayko MM, Meng H, Kleinstein SH, Good-Jacobson KL, and Shlomchik MJ 2014. CD80 and PD-L2 define functionally distinct memory B cell subsets that are independent of antibody isotype. Nature Immunology 15: 631–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Smith KGC, Light A, Nossal GJV, and Tarlinton DM 1997. The extent of affinity maturation differs between the memory and antibody-forming cell compartments in the primary immune response. The EMBO Journal 16: 2996–3006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Dogan I, Bertocci B, Vilmont V, Delbos F, Mégret J, Storck S, Reynaud C-A, and Weill J-C 2009. Multiple layers of B cell memory with different effector functions. Nature Immunology 10: 1292–1299. [DOI] [PubMed] [Google Scholar]
  • 67.Seifert M, Przekopowitz M, Taudien S, Lollies A, Ronge V, Drees B, Lindemann M, Hillen U, Engler H, Singer BB, and Küppers R 2015. Functional capacities of human IgM memory B cells in early inflammatory responses and secondary germinal center reactions. PNAS 112: E546–E555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Shinnakasu R, Inoue T, Kometani K, Moriyama S, Adachi Y, Nakayama M, Takahashi Y, Fukuyama H, Okada T, and Kurosaki T 2016. Regulated selection of germinal-center cells into the memory B cell compartment. Nature Immunology 17: 861–869. [DOI] [PubMed] [Google Scholar]
  • 69.Ise W, Fujii K, Shiroguchi K, Ito A, Kometani K, Takeda K, Kawakami E, Yamashita K, Suzuki K, Okada T, and Kurosaki T 2018. T Follicular Helper Cell-Germinal Center B Cell Interaction Strength Regulates Entry into Plasma Cell or Recycling Germinal Center Cell Fate. Immunity 48: 702–715.e4. [DOI] [PubMed] [Google Scholar]
  • 70.Suan D, Kräutler NJ, Maag JLV, Butt D, Bourne K, Hermes JR, Avery DT, Young C, Statham A, Elliott M, Dinger ME, Basten A, Tangye SG, and Brink R 2017. CCR6 Defines Memory B Cell Precursors in Mouse and Human Germinal Centers, Revealing Light-Zone Location and Predominant Low Antigen Affinity. Immunity 47: 1142–1153.e4. [DOI] [PubMed] [Google Scholar]
  • 71.Hebeis BJ, Klenovsek K, Rohwer P, Ritter U, Schneider A, Mach M, and Winkler TH 2004. Activation of Virus-specific Memory B Cells in the Absence of T Cell Help. J Exp Med 199: 593–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jash A, Wang Y, Weisel FJ, Scharer CD, Boss JM, Shlomchik MJ, and Bhattacharya D 2016. ZBTB32 Restricts the Duration of Memory B Cell Recall Responses. J. Immunol. 197: 1159–1168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Jash A, Zhou YW, Gerardo DK, Ripperger TJ, Parikh BA, Piersma S, Jamwal DR, Kiela PR, Boon ACM, Yokoyama WM, Hsieh CS, and Bhattacharya D 2019. ZBTB32 restrains antibody responses to murine cytomegalovirus infections, but not other repetitive challenges. Sci Rep 9: 15257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lau D, Lan LY-L, Andrews SF, Henry C, Rojas KT, Neu KE, Huang M, Huang Y, DeKosky B, Palm A-KE, Ippolito GC, Georgiou G, and Wilson PC 2017. Low CD21 expression defines a population of recent germinal center graduates primed for plasma cell differentiation. Science Immunology 2: eaai8153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Graham RL, Donaldson EF, and Baric RS 2013. A decade after SARS: strategies for controlling emerging coronaviruses. Nature Reviews Microbiology 11: 836–848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Bradburne AF, Bynoe ML, and Tyrrell DA 1967. Effects of a “new” human respiratory virus in volunteers. Br Med J 3: 767–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Gorse GJ, Patel GB, Vitale JN, and O’Connor TZ 2010. Prevalence of Antibodies to Four Human Coronaviruses Is Lower in Nasal Secretions than in Serum. Clin. Vaccine Immunol. 17: 1875–1880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Chan KH, Cheng VCC, Woo PCY, Lau SKP, Poon LLM, Guan Y, Seto WH, Yuen KY, and Peiris JSM 2005. Serological Responses in Patients with Severe Acute Respiratory Syndrome Coronavirus Infection and Cross-Reactivity with Human Coronaviruses 229E, OC43, and NL63. Clin. Diagn. Lab. Immunol. 12: 1317–1321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Reed SE 1984. The behaviour of recent isolates of human respiratory coronavirus in vitro and in volunteers: Evidence of heterogeneity among 229E-related strains. Journal of Medical Virology 13: 179–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Callow KA, Parry HF, Sergeant M, and Tyrrell D. a. J. 1990. The time course of the immune response to experimental coronavirus infection of man. Epidemiology & Infection 105: 435–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Huang AT, Garcia-Carreras B, Hitchings MDT, Yang B, Katzelnick L, Rattigan SM, Borgert B, Moreno C, Solomon BD, Rodriguez-Barraquer I, Lessler J, Salje H, Burke DS, Wesolowski A, and Cummings DAT 2020. A systematic review of antibody mediated immunity to coronaviruses: antibody kinetics, correlates of protection, and association of antibody responses with severity of disease. medRxiv 2020.04.14.20065771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Gaunt ER, Hardie A, Claas ECJ, Simmonds P, and Templeton KE 2010. Epidemiology and Clinical Presentations of the Four Human Coronaviruses 229E, HKU1, NL63, and OC43 Detected over 3 Years Using a Novel Multiplex Real-Time PCR Method. Journal of Clinical Microbiology 48: 2940–2947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Pyrc K, Dijkman R, Deng L, Jebbink MF, Ross HA, Berkhout B, and van der Hoek L 2006. Mosaic Structure of Human Coronavirus NL63, One Thousand Years of Evolution. Journal of Molecular Biology 364: 964–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Woo PCY, Lau SKP, Yip CCY, Huang Y, Tsoi H-W, Chan K-H, and Yuen K-Y 2006. Comparative Analysis of 22 Coronavirus HKU1 Genomes Reveals a Novel Genotype and Evidence of Natural Recombination in Coronavirus HKU1. Journal of Virology 80: 7136–7145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Lau SKP, Lee P, Tsang AKL, Yip CCY, Tse H, Lee RA, So L-Y, Lau Y-L, Chan K-H, Woo PCY, and Yuen K-Y 2011. Molecular Epidemiology of Human Coronavirus OC43 Reveals Evolution of Different Genotypes over Time and Recent Emergence of a Novel Genotype due to Natural Recombination. Journal of Virology 85: 11325–11337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ladner JT, Larsen BB, Bowers JR, Hepp CM, Bolyen E, Folkerts M, Sheridan K, Pfeiffer A, Yaglom H, Lemmer D, Sahl JW, Kaelin EA, Maqsood R, Bokulich NA, Quirk G, Watt TD, Komatsu K, Waddell V, Lim ES, Caporaso JG, Engelthaler DM, Worobey M, and Keim P 2020. Defining the Pandemic at the State Level: Sequence-Based Epidemiology of the SARS-CoV-2 virus by the Arizona COVID-19 Genomics Union (ACGU). medRxiv 2020.05.08.20095935. [Google Scholar]
  • 87.Ng K, Faulkner N, Cornish G, Rosa A, Earl C, Wrobel A, Benton D, Roustan C, Bolland W, Thompson R, Agua-Doce A, Hobson P, Heaney J, Rickman H, Paraskevopoulou S, Houlihan CF, Thomson K, Sanchez E, Shin GY, Spyer MJ, Walker PA, Kjaer S, Riddell A, Beale R, Swanton C, Gandhi S, Stockinger B, Gamblin S, McCoy LE, Cherepanov P, Nastouli E, and Kassiotis G 2020. Pre-existing and de novo humoral immunity to SARS-CoV-2 in humans. bioRxiv 2020.05.14.095414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Cao W-C, Liu W, Zhang P-H, Zhang F, and Richardus JH 2007. Disappearance of antibodies to SARS-associated coronavirus after recovery. N. Engl. J. Med. 357: 1162–1163. [DOI] [PubMed] [Google Scholar]
  • 89.Guo X, Guo Z, Duan C, Chen Z, Wang G, Lu Y, Li M, and Lu J 2020. Long-Term Persistence of IgG Antibodies in SARS-CoV Infected Healthcare Workers. medRxiv 2020.02.12.20021386. [Google Scholar]
  • 90.Tan CW, Chia WN, Qin X, Liu P, Chen MI-C, Tiu C, Hu Z, Chen VC-W, Young BE, Sia WR, Tan Y-J, Foo R, Yi Y, Lye DC, Anderson DE, and Wang L-F 2020. A SARS-CoV-2 surrogate virus neutralization test based on antibody-mediated blockage of ACE2-spike protein-protein interaction. Nat. Biotechnol. online ahead of print. [DOI] [PubMed] [Google Scholar]
  • 91.Seow J, Graham C, Merrick B, Acors S, Steel KJA, Hemmings O, O’Bryne A, Kouphou N, Pickering S, Galao R, Betancor G, Wilson HD, Signell AW, Winstone H, Kerridge C, Temperton N, Snell L, Bisnauthsing K, Moore A, Green A, Martinez L, Stokes B, Honey J, Izquierdo-Barras A, Arbane G, Patel A, OConnell L, Hara GO, MacMahon E, Douthwaite S, Nebbia G, Batra R, Martinez-Nunez R, Edgeworth JD, Neil SJD, Malim MH, and Doores K 2020. Longitudinal evaluation and decline of antibody responses in SARS-CoV-2 infection. medRxiv 2020.07.09.20148429. [Google Scholar]
  • 92.Falsey AR, Singh HK, and Walsh EE 2006. Serum antibody decay in adults following natural respiratory syncytial virus infection. Journal of Medical Virology 78: 1493–1497. [DOI] [PubMed] [Google Scholar]
  • 93.Lee FE-H, Walsh EE, Falsey AR, Betts RF, and Treanor JJ 2004. Experimental infection of humans with A2 respiratory syncytial virus. Antiviral Research 63: 191–196. [DOI] [PubMed] [Google Scholar]
  • 94.Beem M, Egerer R, and Anderson J 1964. Respiratory Syncytial Virus Neutralizing Antibodies in Persons Residing in Chicago, Illinois. Pediatrics 34: 761–770. [PubMed] [Google Scholar]
  • 95.Habibi MS, Jozwik A, Makris S, Dunning J, Paras A, DeVincenzo JP, de Haan CAM, Wrammert J, Openshaw PJM, and Chiu C 2015. Impaired Antibody-mediated Protection and Defective IgA B-Cell Memory in Experimental Infection of Adults with Respiratory Syncytial Virus. Am J Respir Crit Care Med 191: 1040–1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Hall CB, Geiman JM, Biggar R, Kotok DI, Hogan PM, and Douglas RG 1976. Respiratory Syncytial Virus Infections within Families. New England Journal of Medicine 294: 414–419. [DOI] [PubMed] [Google Scholar]
  • 97.Falsey AR, and Walsh EE 2000. Respiratory Syncytial Virus Infection in Adults. Clinical Microbiology Reviews 13: 371–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Yu X, Tsibane T, McGraw PA, House FS, Keefer CJ, Hicar MD, Tumpey TM, Pappas C, Perrone LA, Martinez O, Stevens J, Wilson IA, Aguilar PV, Altschuler EL, Basler CF, and J. E. C. Jr. 2008. Neutralizing antibodies derived from the B cells of 1918 influenza pandemic survivors. Nature 455: 532–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Vesikari T, Pellegrini M, Karvonen A, Groth N, Borkowski A, O’Hagan DT, and Podda A 2009. Enhanced Immunogenicity of Seasonal Influenza Vaccines in Young Children Using MF59 Adjuvant. The Pediatric Infectious Disease Journal 28: 563–571. [DOI] [PubMed] [Google Scholar]
  • 100.Barclay WS, al-Nakib W, Higgins PG, and Tyrrell DA 1989. The time course of the humoral immune response to rhinovirus infection. Epidemiol Infect 103: 659–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ladner JT, Larsen BB, Bowers JR, Hepp CM, Bolyen E, Folkerts M, Sheridan K, Pfeiffer A, Yaglom H, Lemmer D, Sahl JW, Kaelin EA, Maqsood R, Bokulich NA, Quirk G, Watt TD, Komatsu K, Waddell V, Lim ES, Caporaso JG, Engelthaler DM, Worobey M, and Keim P 2020. Defining the Pandemic at the State Level: Sequence-Based Epidemiology of the SARS-CoV-2 virus by the Arizona COVID-19 Genomics Union (ACGU),. Infectious Diseases (except HIV/AIDS). [Google Scholar]
  • 102.Mulligan MJ, Lyke KE, Kitchin N, Absalon J, Gurtman A, Lockhart SP, Neuzil K, Raabe V, Bailey R, Swanson KA, Li P, Koury K, Kalina W, Cooper D, Fonter-Garfias C, Shi P-Y, Tuereci O, Tompkins KR, Walsh EE, Frenck R, Falsey AR, Dormitzer PR, Gruber WC, Sahin U, and Jansen KU 2020. Phase 1/2 Study to Describe the Safety and Immunogenicity of a COVID-19 RNA Vaccine Candidate (BNT162b1) in Adults 18 to 55 Years of Age: Interim Report. medRxiv 2020.06.30.20142570. [Google Scholar]
  • 103.Nikolich-Zugich J, Knox KS, Rios CT, Natt B, Bhattacharya D, and Fain MJ 2020. SARS-CoV-2 and COVID-19 in older adults: what we may expect regarding pathogenesis, immune responses, and outcomes. GeroScience 42: 505–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mathew D, Giles JR, Baxter AE, Greenplate AR, Wu JE, Alanio C, Oldridge DA, Kuri-Cervantes L, Pampena MB, D’Andrea K, Manne S, Chen Z, Huang YJ, Reilly JP, Weisman AR, Ittner CAG, Kuthuru O, Dougherty J, Nzingha K, Han N, Kim J, Pattekar A, Goodwin EC, Anderson EM, Weirick ME, Gouma S, Arevalo CP, Bolton MJ, Chen F, Lacey SF, Hensley SE, Apostolidis S, Huang AC, Vella LA, Unit UPCP, Betts MR, Meyer NJ, and Wherry EJ 2020. Deep immune profiling of COVID-19 patients reveals patient heterogeneity and distinct immunotypes with implications for therapeutic interventions. bioRxiv 2020.05.20.106401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wertheimer AM, Bennett MS, Park B, Uhrlaub JL, Martinez C, Pulko V, Currier NL, Nikolich-Zugich D, Kaye J, and Nikolich-Zugich J 2014. Aging and cytomegalovirus infection differentially and jointly affect distinct circulating T cell subsets in humans. J Immunol 192: 2143–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Nikolich-Zugich J 2018. The twilight of immunity: emerging concepts in aging of the immune system. Nat Immunol 19: 10–19. [DOI] [PubMed] [Google Scholar]
  • 107.Ni L, Ye F, Cheng ML, Feng Y, Deng YQ, Zhao H, Wei P, Ge J, Gou M, Li X, Sun L, Cao T, Wang P, Zhou C, Zhang R, Liang P, Guo H, Wang X, Qin CF, Chen F, and Dong C 2020. Detection of SARS-CoV-2-Specific Humoral and Cellular Immunity in COVID-19 Convalescent Individuals. Immunity 52: 971–977 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Vabret N, Britton GJ, Gruber C, Hegde S, Kim J, Kuksin M, Levantovsky R, Malle L, Moreira A, Park MD, Pia L, Risson E, Saffern M, Salome B, Esai Selvan M, Spindler MP, Tan J, van der Heide V, Gregory JK, Alexandropoulos K, Bhardwaj N, Brown BD, Greenbaum B, Gumus ZH, Homann D, Horowitz A, Kamphorst AO, Curotto de Lafaille MA, Mehandru S, Merad M, Samstein RM, and P. Sinai Immunology Review. 2020. Immunology of COVID-19: Current State of the Science. Immunity 52: 910–941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.McElhaney JE 2009. Prevention of infectious diseases in older adults through immunization: the challenge of the senescent immune response. Expert Rev Vaccines 8: 593–606. [DOI] [PubMed] [Google Scholar]
  • 110.Lal H, Cunningham AL, Godeaux O, Chlibek R, Diez-Domingo J, Hwang SJ, Levin MJ, McElhaney JE, Poder A, Puig-Barbera J, Vesikari T, Watanabe D, Weckx L, Zahaf T, Heineman TC, and Z. O. E. S. Group. 2015. Efficacy of an adjuvanted herpes zoster subunit vaccine in older adults. N Engl J Med 372: 2087–96. [DOI] [PubMed] [Google Scholar]

RESOURCES