Summary
The brutal toll of another viral pandemic can be blunted by investing now in research that uncovers mechanisms of broad-based immunity so we may have vaccines and therapeutics at the ready. We do not know exactly what pathogen may trigger the next wave or next pandemic. We do know, however, that the human immune system must respond, and must be bolstered with effective vaccines and other therapeutics to preserve lives and livelihoods. These countermeasures must focus on features conserved among families of pathogens in order to be responsive against something yet to emerge. Here, we focus on immunological approaches to mitigate the impact of the next emerging virus pandemic by developing vaccines that elicit both broadly protective antibodies and T cells. Identifying human immune mechanisms of broad protection against virus families with pandemic potential will be our best defense for humanity in the future.
INTRODUCTION
Entering the third year of the COVID pandemic underscores the growing cost of a new infectious agent, whether measured in lives (the staggering number of deaths and long-term complications), or in livelihoods (the toll on education, mental health, businesses and economies). Indeed, the cost of the 2014-2016 Ebola virus epidemic has been estimated at $53 Billion, and that of the SARS-CoV-2 pandemic as $16 Trillion, or 90% of the gross domestic product of the United States. The direct and indirect toll of emerging viruses screams the need for global strategies to anticipate and counteract future pandemics.
Strategies are needed at different levels, from infrastructure and rapid response capabilities to interventions such as broad-based antivirals, monoclonal antibodies, and vaccines. Strategies that induce the broadest possible immunity provide our best hope against as-yet-unknown threats.
In this perspective we focus on broad-based immunity against viruses with pandemic human potential, especially those with potential for zoonotic spillover and emergence of new species of concern. Although global pandemics have this far been caused by influenza viruses (Orthomyxoviridae) and coronaviruses (Coronaviridae), emergence of new species of concern is possible from other viral families, as well including Arenaviridae, Flaviviridae, Bunyavirales, Paramyxoviridae, Togaviridae, Picornaviridae and Filoviridae. Well-known viral species of human health concern from these families include influenza, Lassa, West Nile, Dengue, Nipah/Hendra, Zika, Ebola, SARS, MERS and many others. While we do not know which viral species or variant will next emerge, we do know that the human immune system will need to respond. A focus on human immune mechanisms of broad protection against these families of viruses provides our best defense for humanity in the future.
Here we focus on immunological approaches to blunt the impact of another emerging virus pandemic, namely, development of vaccine concepts aimed at eliciting broadly protective antibodies and T cells alike.
Broadly protective antibodies
Antibodies that achieve protection against multiple viruses must first achieve breadth of recognition. Three points are relevant here. First, although an immune response is polyclonal, mutagenic substitution that reduces recognition of multiple individual antibodies at immunodominant sites will diminish antibody protection: a broadly protective vaccine must elicit antibodies able to recognize an array of mutagenic substitutions. Second, although antiviral antibodies may target a linear sequence or loop, more often, they target a three-dimensional surface conformational in nature, with contributions from amino acids that approach each other in the three-dimensional fold (VanBlargan et al., 2016); Crowe, 2017). In immunogen design, we must consider three-dimensional space, and transient conformational adjustment in that space. Mutations don’t just alter a single binding contact, they can also influence 3D conformation and transient occupancy of different conformers. Third, to target heavily glycosylated viral envelope proteins, antibodies must reach in between carbohydrates or successfully incorporate conserved parts of the carbohydrate into their epitopes (Nguyen et al., 2019); (Hastie et al., 2019; LaBranche et al., 2018); (Dang et al., 2021) (Figure 1A). Some rare antibodies recognize the carbohydrate structure themselves by detecting forms and clusters of carbohydrate unique to a viral surface (Acharya et al., 2020; Saunders et al., 2017; Williams et al., 2021), sometimes even achieving glycan-directed recognition of disparate viral families (Lee et al., 2021). The glycans themselves may not only be essential for recognition by assisting in presentation of the protein structure and conformation. As a result, we must consider glycosylation in immunogen design. Different strategies are needed at different sites. We can block highly variable or undesired antibody sites by adding glycosylation (Chen et al., 2021; Duan et al., 2018; Lin et al., 2021). We can encourage recognition of desired sites by ensuring incorporation of native glycosylation that reflects that of the authentic virus (properly structured antigens). Further, at some difficult to access sites, we can consider deleting glycan in priming steps to facilitate recognition by initial germline antibodies, boosting later with fully and natively glycosylated immunogens (Dubrovskaya et al., 2019; Hastie et al., 2019; LaBranche et al., 2018).
Figure 1: Approaches to the induction of broad-based immunity.
A) To achieve breadth of antibody recognition, vaccine immunogens must faithfully display conformational, heavily glycosylated surfaces and conserved sites, perhaps assisted by multivalent antigen display. Fc-mediated protection should be sought in addition to neutralization. B) Inclusion in the vaccine design of antigens and epitopes from conserved regions of the SARS-CoV-2 genome outside spike could provide additional more broadly cross reactive T cell responses. Image credit: Christina Corbaci.
Most importantly, we must target sites that are conserved in sequence in order to achieve breadth of recognition. Target sites conserved in sequence will frequently be those portions of the envelope protein essential for viral entry, which cannot tolerate mutagenic substitution without a cost to fitness (Figure 1A). For example, stalk, membrane-proximal and stem-helix epitopes contain machinery essential for viral infection and are comparatively conserved in sequence (Guthmiller et al., 2022); (Cagigi et al., 2018; Caillat et al., 2021; Darricarrère et al., 2021; Flyak et al., 2018; King et al., 2019; Li et al., 2022; Pinto et al., 2019; Wang et al., 2021a; Zhou et al., 2022) antibodies against them achieve broad recognition. Other broadly reactive antibodies, for example those that cross react between seasonal coronaviruses and SARS-CoV-2, target sites on the more conserved S2 subunit (Dowell et al., 2022; Ng et al., 2021) rather than the receptor-binding domain. Broad recognition may also occur by contacts to main chain instead of side chain, or to the unchanging carbohydrate cores when the presence of the carbohydrate itself is essential for proper fold of the viral protein.
One key target of broadly reactive antibodies are the hydrophobic fusion loops or fusion peptides indispensable for interaction with and fusion of virus and host membranes (Cheng et al., 2019; Kim et al., 2021; Sauer et al., 2021; Vanderheijden et al., 2021). Not only are these sites more conserved in sequence, protein-protein recognition mediated by hydrophobic surfaces may be more tolerant of side chain substitution than other epitopes that rely on a particular polar hydrogen bond. Antibodies that broadly react with the multiple viruses in the ebolavirus family, for example, recognize the hydrophobic fusion loop largely conserved among the Ebola, Sudan, Bundibugyo, Reston and Tai Forest viruses. These viral glycoproteins are otherwise only ~50% conserved overall (Milligan et al., 2019, 2022; Milligan, J.C., Davis, C.W., Yu, X., Ilinykh, P.A., Huang, K., Halfmann, P., Cross, R.W., Borisevich, V., Agans, K.N., Geisbert,J.B., Chennareddy, C., Goff, A.J., Piper, A.E., Hui, S., Shaffer, K., Buck,T., Heinrich,M.L., Branco, L.M., Crozier, I., Holbrook, M.R., Kuhn, J.H., Kawaoka, Y., Glass,P.J., Bukreyev, A., Geisbert, T.W., Worwa, G., Ahmed,R., and Saphire, E.O., 2022; West et al., 2018). Antibodies against the fusion peptides of previously endemic coronaviruses cross-react with that of SARS-CoV-2 (Vanderheijden et al., 2021).
Another conserved target could be a receptor-binding site shared among multiple viruses in the family, such as those antibodies able to neutralize both SARS-CoV-1 and SARS-CoV-2 by interfering with binding of the shared ACE2 receptor (Jennewein et al., 2021), the broad recognition of influenza viruses by recognition of the shared sialylated glycan receptors (Whittle et al., 2011; Wu and Wilson, 2020); (Lin et al., 2018). Broad recognition of ebolaviruses and even Marburg viruses can occur by recognition of the common NPC1 receptor-binding site (Gong et al., 2016; Hashiguchi et al., 2015). Note however, that the ebolaviruses haven’t changed much in sequence during or between outbreaks: The Ebola virus that broke out in 1976 in the Democratic Republic of the Congo is 99% identical to the Ebola virus that broke out in 2014 in DRC (Jun et al., 2015), and antibodies neutralize both equally well. In contrast, the SARS-CoV-2 pandemic occurred with intense evolution in the virus as it adapted to its new human host. Adaptation through mutations in its receptor binding motif meant a significant drop in binding of many anti-receptor binding site antibodies to the beta and omicron variants (Dejnirattisai et al., 2022; Wang et al., 2021b). Vaccination strategies against rapidly evolving viral sequences, for SARS-CoV-2 or other viruses, will need to involve a variety of sequences (i.e. display of antigen or portions of antigen from multiple members of the virus family), or will need to better display or prime with conserved, perhaps less immunodominant, portions of the envelope proteins conserved among members of the virus family (Cheng et al., 2019; Cottrell et al., 2020).
Breadth of recognition and potency of neutralization is often achieved by recognition of quaternary epitopes, formed when multiple subunits of the viral surface proteins approach each other in three-dimensional space (de Alwis et al., 2012; Dang et al., 2021; Tyagi et al., 2020). Some of the more potently neutralizing antibodies against multiple families of viruses recognize multiple subunits of the viral glycoprotein, whether by bridging the receptor-binding subunit to the fusion subunit, by anchoring neighboring monomers in in a single glycoprotein trimer together, or by bridging multiple copies of envelope on the viral surface (de Alwis et al., 2012; Fedechkin et al., 2020). Vaccine strategies that elicit these types of antibodies must faithfully display the right conformation and the right assembly of these unstable proteins – tactics which require protein engineering of the immunogen to achieve.
Creation of an antibody protein surface (paratope) that achieves precise recognition of a novel pathogen is achieved over time, evolving from initial, low-affinity germline sequences, to higher-affinity binding molecules. Repeated antigen stimulation to evolve these responses is facilitated by longevity/stability of the immunogen, boosting or exposure (Tsai et al., 2013). In design of vaccine strategies, we have the opportunity to prime with one form and boost with another to improve more elusive, but desired types of antibody responses.
Geometry and stoichiometry matter here: in general, recognition of repeated copies of antigen facilities binding by avidity (Jendroszek and Kjaergaard; Oda and Azuma, 2000; Remmel et al., 2021; Yin et al.) (Figure 1A). Recognition by the natural IgG may be monovalent, bivalent, or may occur by bridging separate glycoproteins together. In vaccine design, display of antigen in the correct oligomeric form and in desired immunogen spacing will assist in elicitation of IgG that naturally bridge monomers in the trimer, that bind with avidity (Tsai et al., 2013), and which display Fc for immune effector cells. Multivalent display of antigens on vaccine particles (whether nanoparticles or virus-like particles or the surfaces of attenuated viruses), and display of full oligomeric glycoproteins instead of single subunits, will allow vaccines to elicit IgG that can take advantage of oligomeric state and geometry to achieve both neutralizing and extra-neutralizing functions. In one broad analysis of an array of therapeutic antibodies against SARS-CoV-2, those antibodies that take advantage of avidity achieve more potent neutralization and breadth of recognition than those that engage by one Fab alone (Hastie et al., 2021). Elicitation of these types of antibodies would be facilitated by display of whole glycoprotein oligomers, rather than monomeric receptor-binding domains. Further, display of multiple copies on nanoparticles would afford opportunities to enhance responses against desired, conserved, otherwise weakly immunogenic epitopes (Yang et al., 2020). For viruses like Dengue, one must consider whether the particles are mature: better neutralization results may be achieved when particle preparations are homogeneous and mature (Tsai et al., 2018; Yang et al., 2020), presenting conformations and quaternary sites accessible in the actual infecting virion.
In understanding which are the desirable types of antibodies to elicit with a vaccine, we must note that there are both neutralizing and extra-neutralizing functions of antibodies. Neutralization means the ability of antibody to prevent viral propagation in cell culture, usually of a single cell line. Neutralization in vitro is usually mechanical in nature and involves the biochemistry and biophysics of the protein-protein interactions between antibody and pathogen. Antibody binding that sterically blocks binding of receptor, required protease cleavage of the viral glycoprotein, or conformational changes required for fusion of viral and host membranes will all achieve mechanical neutralization. Neutralization is measured in vitro by measuring viral or pseudoviral infection of cells in culture, often using a GFP- or other reporter gene to measure infection. Because neutralization is easy to measure, and peripheral serum antibody easy to collect, neutralization of serum antibody is a frequently tapped tool to forecast vaccine efficacy. Note also that mechanical neutralization, which involves the biochemical and biophysical events of protein-protein interaction, is more straightforward to recapitulate among tissue culture, rodents, non-human primates and human clinical studies.
However, although neutralization in vitro is a key predictor, it is not the only predictor of antibody efficacy in vivo (Balachandran et al., 1982; Calvert et al., 2022; Corbeil et al., 1996; Kim et al., 2021). Some antibodies have a mechanical activity not recapitulated in the chosen in vitro system, depending, for example, on whether the range of target cells in vivo has different conditions or entry factors than the cell type used in vitro (Neerukonda et al., 2021). Some protective antibodies block egress instead of entry (Williamson et al., 2021); (Dufloo et al., 2022; Jin et al., 2018).
Other antibodies recruit protection in vivo via their Fc domain. In this process, antibody-bound particles bind to and trigger Fc receptors, which result in the generation, secretion or repression of various pro- or anti-inflammatory substances. Fc-mediated immune effector functions are a major contributor of antibody-mediated protection, whether by passive immunotherapy or by vaccines, and are a fundamental source of antibody protection. While the activity of the Fab may inactivate a virus, it is the activity of the Fc that clears the virus and infected cells from the body. These Fc-mediated antibody functions (Forthal, 2014); (Bournazos and Ravetch, 2017) involve lysis of infected cells, immune activation by complement, internalization and destruction of virions and infected cells by phagocytosis, modulation of inflammation, mucus trapping, and recruitment of other factors or cells in the immune system. Fc-mediated internalization of immune complexes results in the engagement of toll-like receptors. Further, Fc-mediated phagocytosis of foreign particles by antigen- presenting cells enhances T cell responses. Hence, achieving extra-neutralizing functions can assist development of broadly reactive and broadly protective T cell responses.
Fc functions occur outside of and independently from in vitro neutralization. They may occur on neutralizing antibodies or on non-neutralizing antibodies. Hence, we prefer to term them “extra-neutralization” functions rather than non-neutralizing functions, because “non-neutralizing” implies that these functions only occur on antibodies that do not neutralize. Instead, Fc-mediated protective functions may occur in the presence or absence of complementary neutralization. Indeed, addition of Fc-mediated functions on top of Fab-mediated neutralization provides additional protective benefit (Hessell et al., 2007) over mechanical neutralization alone.
These extra-neutralization functions are measured less frequently than neutralization as they are more difficult to measure, involving multiple factors and multiple cell types, concerted and sometimes transient or low-affinity interactions. Further, Fc-mediated functions differ among each animal model species and humans. These extra-neutralization functions are, however, important. They have been linked to improved survival in viral infection (Bahnan et al., 2021; Hessell et al., 2007), and may constitute the majority of the antibody response: The neutralizing portion of antibodies, for example, are a fraction of the total antibody elicited in SARS-CoV-2 infection (Yamayoshi et al., 2021), and for Ebola virus, total binding antibody is a stronger correlate of protection than neutralizing antibody (Roozendaal et al., 2020).
Interestingly, in some studies, it is the broadly reactive antibodies that achieve the greatest Fc-mediated effect (Terajima et al., 2011); (Grunst and Uchil, 2022). Hence, antibodies that broadly bind can be protective, even if they do not neutralize (van Erp et al., 2019); (Thulin and Wang, 2018). For coronaviruses, for breadth, one may seek to target the S2 subunit (Dowell et al., 2022); (Pinto et al., 2021), a subunit characterized by much lower potency of neutralization. Vaccines against other families of viruses should seek recognition of more conserved subunits as well (Angeletti and Yewdell, 2018; Ellebedy et al., 2014; Hsieh et al., 2021; Impagliazzo et al., 2015; Mallajosyula et al., 2014). Such antibodies may provide protection in humans even if they do not lead to the most potent mechanical neutralization in cell culture.
The low-hanging fruit, or straightforward way to achieve broad recognition may simply be including antigens from different viral strains or species onto one vaccine particle, for example the receptor-binding domains from multiple coronaviruses (Dowell et al., 2022; Walls et al., 2021) or the envelopes of multiple ebolaviruses (Callendret et al., 2018; Cross et al., 2020; Lehrer et al., 2021; Matassov et al., 2018). The multi-component strategy achieves breadth at the polyclonal level, eliciting some antibodies against each of the components individually and some antibodies that target something shared among them. The high-hanging fruit, identification of immunogens that represent shared features of the multiple strains or viruses in a family, involves engineering of viral antigens to mask immunodominant, but variable sites, and to better stabilize and present conserved and essential, but less immunodominant sites. The vaccines in the SARS-CoV-2 pandemic could be rapidly mobilized because of the prior decades of vaccine research into engineering strategies, display formats, and related viruses. To blunt the impact of the next pandemic, we must continue exploration of vaccine strategies against the array of virus families, seeking to elicit antibodies that are broad in recognition, and broad in types of immune protection achieved. In addition to seeking the most potent mechanical neutralization, we must also think more deeply about engineering immunogens that faithfully present quaternary assembly and glycosylation of the actual virus, correct multimeric presentation to take advantage of beneficial effects of avidity. We must also seek better methods of measuring and encourage the multitude of protective Fc-mediated functions for both clearance and inspiration of complementary T cell activities.
Broadly protective T cells
What is the rationale for the further exploration of a broadly cross-reactive T cell vaccine concept? To illustrate this point we briefly point to some of the lines of evidence supporting the concept in the case of SARS-CoV-2. Early T cell responses correlate with better outcomes (Rydyznski Moderbacher et al., 2020) and lower viral loads (Tan et al., 2021). Agammaglobulinemic and B cell-depleted individuals have only moderately increased risk of hospitalization with COVID-19 (Soresina et al., 2020); COVID-19 in ocrelizumab-treated people with multiple sclerosis (MS) is predominantly mild, suggesting that disease can be controlled and resolved also in absence of antibody responses (Apostolidis et al., 2021; Sabatino et al., 2022). The onset of protection following one-dose of Moderna or Pfizer vaccine correlates with early appearance of T cells responses, with detectable neutralizing antibodies (Kalimuddin et al., 2021). Thus, several lines of evidence point to a potential protective contribution of T cells in the context of control of SARSCoV-2 and modulation of disease severity. Additional lines of evidence are provided from studies linking preexisting cross-reactive T cell immunity with vaccination and infection outcomes, discussed in a separate section below.
SARS-CoV-2 variants have taken center stage in the pandemic developments in late 2021 and early 2022. In general, several variants such as Beta, Delta and Omicron have been associated with increased capacity to evade humoral responses induced by natural infection and vaccination, and have also been associated with higher infectivity and transmissibility. However, it has also been noted that in general the variants have not been associated with more severe disease, and in fact Omicron appears to be associated with relatively less severe disease outcomes. The mechanisms associated with these effects are likely to be diverse and factors such as significant changes in Omicron virulence, memory B cells making anamnestic neutralizing antibody responses and changes in tropism infectivity are all plausible contributing factors.
Several studies have investigated whether the T cell response of vaccinated or naturally infected individuals is impaired by variant-associated mutations. The general consensus is that while the mutations affect some epitopes, the majority of T cell epitopes are fully conserved in the various variants (Tarke et al., 2021a); (Redd et al., 2021; Tarke et al., 2021b). Likewise, at the population level it was found that T cell responses are largely preserved in terms of recognition of variants, including Delta and Omicron (Tarke et al., 2021a); (Tarke et al., 2022); (Geers et al., 2021); (Alter et al., 2021); (Alter et al., 2021; Keeton et al., 2021); (Melo-González et al., 2021); (Madelon et al., 2021); (GeurtsvanKessel et al., 2022); (GeurtsvanKessel et al., 2022; Keeton et al., 2022); (De Marco et al., 2021); (Liu et al., 2022); (Gao et al., 2022). In light of this data, it is plausible that T cells play a role in protection, modulation of disease severity and termination of infection. These data suggest that preservation of T cell responses can be of potential benefit, even in concomitance with suboptimal antibody responses.It was noted early on that memory T cell reactivity is also detected in non-exposed individuals (Grifoni et al., 2020a). It was demonstrated (Mateus et al., 2020) that this was to be ascribed to memory T cells recognizing Common Cold Coronavirus (CCC) epitopes with sequence homology to SARS-CoV-2 sequences, although potential crossreactivity of T cell epitopes derived from other viral species was also reported (Bacher et al., 2020); (Bacher et al., 2020; Le Bert et al., 2021). Pre-existing memory T cells in SARS-CoV-2 unexposed subjects capable of recognizing SARS-CoV-2 sequences was reported by several independent parallel studies (reviewed in (Sette and Crotty, 2020). Thus, this observation was independently reported by several groups in diverse settings and geographical locations. Furthermore, numerous studies reported different epitopes associated with different degrees of CCC-SARS-CoV-2 cross-reactivity (Schulien et al., 2021); (Sekine et al., 2020); (Shomuradova et al., 2020); (Nelde et al., 2021); (Keller et al., 2020; Nelde et al., 2021); (Ferretti et al., 2020); (Ferretti et al., 2020; Prakash et al., 2021).
Several additional studies reported cross-reactivity between SARSCoV-2, SARS-CoV-1 and MERS at the level of T cell responses. In general, the level of cross-reactivity tends to be higher when these viruses are considered as compared to CCC, which is consistent with the closer phylogenetic relation, and the higher degree of structural similarity and sequence homology (Ferretti et al., 2020; Habel et al., 2020; Le Bert et al., 2020; Prakash et al., 2021).
It was hypothesized that this pre-existing immunity could influence the severity of disease associated with subsequent SARS-CoV-2 infection and/or the outcomes of SARS-CoV-2 vaccination (Sette and Crotty, 2020). Our recent studies analyzing responses to low dose Moderna mRNA-1273 COVID-19 vaccine demonstrated that indeed subjects with pre-existing cross-reactive immunity respond faster and better at the level of CD4+, T folicular helper (Tfh) and serological responses (Mateus et al., 2021).
Further evidence of cross-reactive T cells roles in prevention of symptomatic COVID-19 was also published by Thiel and colleagues (Loyal et al., 2021). Also consistent with this notion are studies showing a potential protective effect of a conserved HLA-B7 restricted epitope (Peng et al., 2021) and the ability of T cell to cross react with CCC homologous peptides in unexposed individuals (Lineburg et al., 2021). Further studies from Sagar et al showed that recent exposure to common cold coronaviruses correlated with less severe COVID-19 outcomes (Sagar et al., 2021). Finally, we (da Silva Antunes et al., 2021) demonstrated that heavily exposed COVID seronegative donors had high level of CCC-T cell reactivity, and a recent study from the Maini and Bertoletti’s group highlighted that cross-reactive T cells are associated with abortive SARS-CoV-2 infection of health care workers (Swadling et al., 2022). Similarly, a study from Lalvani’s group associated cross-reactive memory T cells with protection against SARS-CoV-2 infection in COVID-19 contacts (Kundu et al., 2022). Thus, cross-reactive T cells can modulate disease severity and dramatically influence infection outcomes (Peng et al., 2021).
The data and studies described above indicate that it might be possible to define antigens, antigen fragments and/or epitopes that are broadly cross-reactive different coronaviruses (Mateus et al., 2020; Prakash et al., 2021)(Fig 1B). Beyond SARS-CoV-1, SARS-CoV-2 and MERS, several other coronaviruses are also of concern, and in particular several zoonotic viruses that could trigger a new pandemic if capable of jumping to human host and causing widespread disease, or even SARS-CoV-2 variants that further evolved in non-human hosts. The identification of cross-reactive epitopes can be accomplished utilizing a combination of immunological, virological and phylogenetic analyses. For example, representative viruses sampling the viral species of concern can be defined, and the sequence conservation along the proteome can be rigorously defined, both within different viral groups and across more distantly related viral groups.
A variety of different immunological techniques can be utilized (Sidney et al., 2020) to define the actual T cell epitopes recognized in individuals infected by CCC or SARS-CoV-2. The conservation and immunogenicity analysis can then be further integrated to map immunogenic and conserved regions in CCC and SARS-CoV-2, and experimentally determining epitopes/ regions that are widely cross-reactive at the level of T. cell responses (Grifoni et al., 2020b; Mateus et al., 2020). The widely cross-reactive antigens, antigen domains and/or epitopes identified could be used as a T cell-inducing vaccine component, either standing alone or in conjunction with antibody-induced components (Dangi et al., 2021).
The definition of conserved/cross-reactive T cell epitopes across different coronaviruses is not without precedent. Cross-reactive and conserved epitopes have been described in a number of different viral species, serotypes and subtypes. Here, as a way of illustration we describe three such instances that we have addressed in past investigations.
In the case of influenza, early studies demonstrated that the human T cell repertoire is broad and multispecific (Gianfrani et al., 2000). A subsequent study described 38 class I and 16 class II nonredundant epitopes which were consistently recognized in multiple subjects, provided high coverage among different ethnicities, and were conserved in the majority of the strains analyzed (Assarsson et al., 2008). Parallel studies focused on HLA DR restricted epitopes conserved in Influenza A virus strains in circulation, associated with past pandemics and of potential zoonotic interest. Immunization of HLA transgenic mice with a DNA plasmid encoding 20 different epitopes was shown to enhance antibody responses and protect from lethal PR8 influenza virus challenge (Alexander et al., 2010a). A similar study studied HLA class I restricted influenza-derived conserved T cell epitopes (Alexander et al., 2010b). Finally, we also reported pre-existing immunity against swine-origin H1N1 influenza viruses in the general human population, presumably due to previous exposure to related influenza strains, and hypothesized that this effect might explain the observed lower disease severity in older subjects, presumably exposed to other H1N1 strains (Greenbaum et al., 2009). Similar approaches were utilized to define sets of epitopes immunogenic in humans and/or restricted by human HLA, and conserved in Old and New World Arenaviruses (Botten et al., 2010; Kotturi et al., 2009, 2010).
A significant amount of data is also available in the case of flaviviruses. In the case of DENV, we have shown that infection with multiple serotypes tend to favor a bias towards recognition of T cell epitopes conserved across serotypes (Weiskopf et al., 2014), and vaccination with a tetravalent attenuated DENV vaccine elicits responses mostly directed against conserved epitopes (Weiskopf et al., 2015). HLA alleles association with disease protection also suggest a potential protective role for T cell responses (Weiskopf et al., 2013, 2016). Several animal model studies have shown that T cell responses can protect from challenge or ameliorate disease (Elong Ngono et al., 2016; Yauch et al., 2009, 2010; Zellweger et al., 2015). Further studies have shown that in DENV infection can influence T cell responses to ZIKV upon ZIKV infection (Grifoni et al., 2017; Wen et al., 2017) presumably because of preexisting ZIKA cross-reactive T cells (Schouest et al., 2021). In, general cross-reactivity amongst different flaviviruses (Dengue, Yellow Fever, Zika) is low (Grifoni et al., 2020b), but conserved epitopes can be identified and shown to mediate protection in animal models of disease (Regla-Nava et al., 2018; Wen et al., 2020).
We conclude that it is possible to entertain a broad concept, aimed at generating T cell inducing vaccine components, for the specific purpose of enhance preparedness against future possible pandemics. Such vaccine components could entail inclusion of additional SARS-CoV-2 antigens such a N or some of the NSP antigens encoded in the Orf1a/b region of the genome, or specific antigen domains of fragments rich in T cell epitope content, or collections of defined T cell epitopes. These T cell vaccine or vaccine components might be designed specifically for the purpose of broadly preventing hospitalizations, severe disease and death. Several groups have reported initial testing of SARS-CoV-2 vaccines including additional components, beyond the spike antigen, which would be expected to broaden the spectrum of T cell reactivities elicited by vaccination. These include NantWorks /Immunitbio, Gritstone, Flowpharma, Walz group, Vaxxinity, several academic groups and potentially others.
We emphasize that this concept should not be seen as an alternative to antibody-inducing strategies, but should rather be expected to be highly synergistic with those strategies. A T cell-inducing component could be produced, tested for safety and immunogenicity in small phase I trials, and even stockpiled, as a first line of defense from a new pandemic, buying time until outbreak-specific vaccine or antibodies become available. Some are in development. This would be done in parallel and as an adjuvant to other antibody inducing vaccine strategies.
Finally, we note that while the approach is being considered for coronaviruses in general, and sarbecoviruses in particular, a similar strategy could be developed for each of several families of viruses of pandemic preparedness concern, namely Arenaviridae, Flaviviridae, Bunyavirales, Paramyxoviridae, Togaviridae, Picornaviridae and Filioviridae. Research now into vaccine strategies that elicit broadly reactive antibodies and T cells alike, against the array of possible threats, will better prepare us, our health systems and our economies, for the next pandemic to come.
HIghlights and eTOC.
Saphire and Sette discuss potential approaches for vaccine development against the broad array of SARS-CoV-2 variants and other viruses of pandemic potential. Approaches that elicit both humoral and cellular responses are considered and are expected to be synergistic with each other, providing humanity with the best chance to defend against the next pandemic.
Acknowledgments:
This project has been funded in whole or in part with Federal funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, under grants U19AI142790 and P01AI165072 and contracts 75N93021C00016 and 75N93019C00065, as well as The Bill and Melinda Gates Foundation grant INV-006133.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declaration of interests. A.S. is a consultant for Gritstone Bio, Flow Pharma, ImmunoScape, Moderna, AstraZeneca, Avalia, Fortress, Repertoire, Gilead, Gerson Lehrman Group, RiverVest, MedaCorp, and Guggenheim. LJI has filed for patent protection for various aspects of T cell epitope and vaccine design work.
References:
- Acharya P, Williams W, Henderson R, Janowska K, Manne K, Parks R, Deyton M, Sprenz J, Stalls V, Kopp M, et al. (2020). A glycan cluster on the SARS-CoV-2 spike ectodomain is recognized by Fab-dimerized glycan-reactive antibodies. bioRxiv. [Google Scholar]
- Alexander J, Bilsel P, del Guercio M-F, Stewart S, Marinkovic-Petrovic A, Southwood S, Crimi C, Vang L, Walker L, Ishioka G, et al. (2010a). Universal influenza DNA vaccine encoding conserved CD4+ T cell epitopes protects against lethal viral challenge in HLA-DR transgenic mice. Vaccine 28, 664–672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander J, Bilsel P, del Guercio M-F, Marinkovic-Petrovic A, Southwood S, Stewart S, Ishioka G, Kotturi MF, Botten J, Sidney J, et al. (2010b). Identification of broad binding class I HLA supertype epitopes to provide universal coverage of influenza A virus. Hum. Immunol 71, 468–474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alter G, Yu J, Liu J, Chandrashekar A, Borducchi EN, Tostanoski LH, McMahan K, Jacob-Dolan C, Martinez DR, Chang A, et al. (2021). Immunogenicity of Ad26.COV2.S vaccine against SARS-CoV-2 variants in humans. Nature 596, 268–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Alwis R, Smith SA, Olivarez NP, Messer WB, Huynh JP, Wahala WMPB, White LJ, Diamond MS, Baric RS, Crowe JE Jr, et al. (2012). Identification of human neutralizing antibodies that bind to complex epitopes on dengue virions. Proc. Natl. Acad. Sci. U. S. A 109, 7439–7444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Angeletti D, and Yewdell JW (2018). Is It Possible to Develop a “Universal” Influenza Virus Vaccine? Outflanking Antibody Immunodominance on the Road to Universal Influenza Vaccination. Cold Spring Harb. Perspect. Biol 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Apostolidis SA, Kakara M, Painter MM, Goel RR, Mathew D, Lenzi K, Rezk A, Patterson KR, Espinoza DA, Kadri JC, et al. (2021). Cellular and humoral immune responses following SARS-CoV-2 mRNA vaccination in patients with multiple sclerosis on anti-CD20 therapy. Nat. Med 27, 1990–2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Assarsson E, Bui H-H, Sidney J, Zhang Q, Glenn J, Oseroff C, Mbawuike IN, Alexander J, Newman MJ, Grey H, et al. (2008). Immunomic analysis of the repertoire of T-cell specificities for influenza A virus in humans. J. Virol 82, 12241–12251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bacher P, Rosati E, Esser D, Martini GR, Saggau C, Schiminsky E, Dargvainiene J, Schröder I, Wieters I, Khodamoradi Y, et al. (2020). Low-Avidity CD4 T Cell Responses to SARS-CoV-2 in Unexposed Individuals and Humans with Severe COVID-19. Immunity 53, 1258–1271.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bahnan W, Wrighton S, Sundwall M, Bläckberg A, Larsson O, Höglund U, Khakzad H, Godzwon M, Walle M, Elder E, et al. (2021). Spike-Dependent Opsonization Indicates Both Dose-Dependent Inhibition of Phagocytosis and That Non-Neutralizing Antibodies Can Confer Protection to SARS-CoV-2. Front. Immunol 12, 808932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balachandran N, Bacchetti S, and Rawls WE (1982). Protection against lethal challenge of BALB/c mice by passive transfer of monoclonal antibodies to five glycoproteins of herpes simplex virus type 2. Infect. Immun 37, 1132–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Botten J, Whitton JL, Barrowman P, Sidney J, Whitmire JK, Alexander J, Kotturi MF, Sette A, and Buchmeier MJ (2010). A multivalent vaccination strategy for the prevention of Old World arenavirus infection in humans. J. Virol 84, 9947–9956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bournazos S, and Ravetch JV (2017). Fcγ Receptor Function and the Design of Vaccination Strategies. Immunity 47, 224–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cagigi A, Ploquin A, Niezold T, Zhou Y, Tsybovsky Y, Misasi J, and Sullivan NJ (2018). Vaccine-Mediated Induction of an Ebolavirus Cross-Species Antibody Binding to Conserved Epitopes on the Glycoprotein Heptad Repeat 2/Membrane-Proximal External Junction. J. Infect. Dis 218, S537–S544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caillat C, Guilligay D, Torralba J, Friedrich N, Nieva JL, Trkola A, Chipot CJ, Dehez FL, and Weissenhorn W (2021). Structure of HIV-1 gp41 with its membrane anchors targeted by neutralizing antibodies. Elife 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Callendret B, Vellinga J, Wunderlich K, Rodriguez A, Steigerwald R, Dirmeier U, Cheminay C, Volkmann A, Brasel T, Carrion R, et al. (2018). A prophylactic multivalent vaccine against different filovirus species is immunogenic and provides protection from lethal infections with Ebolavirus and Marburgvirus species in non-human primates. PLoS One 13, e0192312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calvert AE, Bennett SL, Hunt AR, Fong RH, Doranz BJ, Roehrig JT, and Blair CD (2022). Exposing cryptic epitopes on the Venezuelan equine encephalitis virus E1 glycoprotein prior to treatment with alphavirus cross-reactive monoclonal antibody allows blockage of replication early in infection. Virology 565, 13–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen T-H, Yang Y-L, Jan J-T, Chen C-C, and Wu S-C (2021). Site-Specific Glycan-Masking/Unmasking Hemagglutinin Antigen Design to Elicit Broadly Neutralizing and Stem-Binding Antibodies Against Highly Pathogenic Avian Influenza H5N1 Virus Infections. Front. Immunol 12, 692700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng C, Xu K, Kong R, Chuang G-Y, Corrigan AR, Geng H, Hill KR, Jafari AJ, O’Dell S, Ou L, et al. (2019). Consistent elicitation of cross-clade HIV-neutralizing responses achieved in guinea pigs after fusion peptide priming by repetitive envelope trimer boosting. PLoS One 14, e0215163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corbeil S, Seguin C, and Trudel M (1996). Involvement of the complement system in the protection of mice from challenge with respiratory syncytial virus Long strain following passive immunization with monoclonal antibody 18A2B2. Vaccine 14, 521–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cottrell CA, van Schooten J, Bowman CA, Yuan M, Oyen D, Shin M, Morpurgo R, van der Woude P, van Breemen M, Torres JL, et al. (2020). Mapping the immunogenic landscape of near-native HIV-1 envelope trimers in non-human primates. PLoS Pathog. 16, e1008753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cross RW, Xu R, Matassov D, Hamm S, Latham TE, Gerardi CS, Nowak RM, Geisbert JB, Ota-Setlik A, Agans KN, et al. (2020). Quadrivalent VesiculoVax vaccine protects nonhuman primates from viral-induced hemorrhagic fever and death. J. Clin. Invest 130, 539–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crowe JE Jr (2017). Principles of Broad and Potent Antiviral Human Antibodies: Insights for Vaccine Design. Cell Host Microbe 22, 193–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dang HV, Cross RW, Borisevich V, Bornholdt ZA, West BR, Chan Y-P, Mire CE, Da Silva SC, Dimitrov AS, Yan L, et al. (2021). Broadly neutralizing antibody cocktails targeting Nipah virus and Hendra virus fusion glycoproteins. Nat. Struct. Mol. Biol 28, 426–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dangi T, Class J, Palacio N, Richner JM, and Penaloza MacMaster P (2021). Combining spike- and nucleocapsid-based vaccines improves distal control of SARS-CoV-2. Cell Rep. 36, 109664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darricarrère N, Qiu Y, Kanekiyo M, Creanga A, Gillespie RA, Moin SM, Saleh J, Sancho J, Chou T-H, Zhou Y, et al. (2021). Broad neutralization of H1 and H3 viruses by adjuvanted influenza HA stem vaccines in nonhuman primates. Sci. Transl. Med 13. [DOI] [PubMed] [Google Scholar]
- Dejnirattisai W, Huo J, Zhou D, Zahradník J, Supasa P, Liu C, Duyvesteyn HME, Ginn HM, Mentzer AJ, Tuekprakhon A, et al. (2022). SARS-CoV-2 Omicron-B.1.1.529 leads to widespread escape from neutralizing antibody responses. Cell 185, 467–484.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Marco L, D’Orso S, Pirronello M, Verdiani A, Termine A, Fabrizio C, Capone A, Sabatini A, Guerrera G, Placido R, et al. (2021). Preserved T cell reactivity to the SARS-CoV-2 Omicron variant indicates continued protection in vaccinated individuals. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dowell AC, Butler MS, Jinks E, Tut G, Lancaster T, Sylla P, Begum J, Bruton R, Pearce H, Verma K, et al. (2022). Children develop robust and sustained cross-reactive spike-specific immune responses to SARS-CoV-2 infection. Nat. Immunol 23, 40–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duan H, Chen X, Boyington JC, Cheng C, Zhang Y, Jafari AJ, Stephens T, Tsybovsky Y, Kalyuzhniy O, Zhao P, et al. (2018). Glycan Masking Focuses Immune Responses to the HIV-1 CD4-Binding Site and Enhances Elicitation of VRC01-Class Precursor Antibodies. Immunity 49, 301–311.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dubrovskaya V, Tran K, Ozorowski G, Guenaga J, Wilson R, Bale S, Cottrell CA, Turner HL, Seabright G, O’Dell S, et al. (2019). Vaccination with Glycan-Modified HIV NFL Envelope Trimer-Liposomes Elicits Broadly Neutralizing Antibodies to Multiple Sites of Vulnerability. Immunity 51, 915–929.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dufloo J, Planchais C, Frémont S, Lorin V, Guivel-Benhassine F, Stefic K, Casartelli N, Echard A, Roingeard P, Mouquet H, et al. (2022). Broadly neutralizing anti-HIV-1 antibodies tether viral particles at the surface of infected cells. Nat. Commun 13, 630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ellebedy AH, Krammer F, Li G-M, Miller MS, Chiu C, Wrammert J, Chang CY, Davis CW, McCausland M, Elbein R, et al. (2014). Induction of broadly cross-reactive antibody responses to the influenza HA stem region following H5N1 vaccination in humans. Proc. Natl. Acad. Sci. U. S. A 111, 13133–13138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elong Ngono A, Chen H-W, Tang WW, Joo Y, King K, Weiskopf D, Sidney J, Sette A, and Shresta S (2016). Protective Role of Cross-Reactive CD8 T Cells Against Dengue Virus Infection. EBioMedicine 13, 284–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Erp EA, Luytjes W, Ferwerda G, and van Kasteren PB (2019). Fc-Mediated Antibody Effector Functions During Respiratory Syncytial Virus Infection and Disease. Front. Immunol 10, 548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fedechkin SO, George NL, Nuñez Castrejon AM, Dillen JR, Kauvar LM, and DuBois RM (2020). Conformational Flexibility in Respiratory Syncytial Virus G Neutralizing Epitopes. J. Virol 94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferretti AP, Kula T, Wang Y, Nguyen DMV, Weinheimer A, Dunlap GS, Xu Q, Nabilsi N, Perullo CR, Cristofaro AW, et al. (2020). Unbiased Screens Show CD8 T Cells of COVID-19 Patients Recognize Shared Epitopes in SARS-CoV-2 that Largely Reside outside the Spike Protein. Immunity 53, 1095–1107.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flyak AI, Kuzmina N, Murin CD, Bryan C, Davidson E, Gilchuk P, Gulka CP, Ilinykh PA, Shen X, Huang K, et al. (2018). Broadly neutralizing antibodies from human survivors target a conserved site in the Ebola virus glycoprotein HR2-MPER region. Nat Microbiol 3, 670–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Forthal DN (2014). Functions of Antibodies. Microbiol Spectr 2, 1–17. [PMC free article] [PubMed] [Google Scholar]
- Gao Y, Cai C, Grifoni A, Müller TR, Niessl J, Olofsson A, Humbert M, Hansson L, Österborg A, Bergman P, et al. (2022). Ancestral SARS-CoV-2-specific T cells cross-recognize the Omicron variant. Nat. Med [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geers D, Shamier MC, Bogers S, den Hartog G, Gommers L, Nieuwkoop NN, Schmitz KS, Rijsbergen LC, van Osch JAT, Dijkhuizen E, et al. (2021). SARS-CoV-2 variants of concern partially escape humoral but not T-cell responses in COVID-19 convalescent donors and vaccinees. Sci Immunol 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GeurtsvanKessel CH, Geers D, Schmitz KS, Mykytyn AZ, Lamers MM, Bogers S, Scherbeijn S, Gommers L, Sablerolles RSG, Nieuwkoop NN, et al. (2022). Divergent SARS CoV-2 Omicron-reactive T- and B cell responses in COVID-19 vaccine recipients. Sci Immunol eabo2202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gianfrani C, Oseroff C, Sidney J, Chesnut RW, and Sette A (2000). Human memory CTL response specific for influenza A virus is broad and multispecific. Hum. Immunol 61, 438–452. [DOI] [PubMed] [Google Scholar]
- Gong X, Qian H, Zhou X, Wu J, Wan T, Cao P, Huang W, Zhao X, Wang X, Wang P, et al. (2016). Structural Insights into the Niemann-Pick C1 (NPC1)-Mediated Cholesterol Transfer and Ebola Infection. Cell 165, 1467–1478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenbaum JA, Kotturi MF, Kim Y, Oseroff C, Vaughan K, Salimi N, Vita R, Ponomarenko J, Scheuermann RH, Sette A, et al. (2009). Pre-existing immunity against swine-origin H1N1 influenza viruses in the general human population. Proc. Natl. Acad. Sci. U. S. A 106, 20365–20370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grifoni A, Pham J, Sidney J, O’Rourke PH, Paul S, Peters B, Martini SR, de Silva AD, Ricciardi MJ, Magnani DM, et al. (2017). Prior Dengue Virus Exposure Shapes T Cell Immunity to Zika Virus in Humans. J. Virol 91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grifoni A, Weiskopf D, Ramirez SI, Mateus J, Dan JM, Moderbacher CR, Rawlings SA, Sutherland A, Premkumar L, Jadi RS, et al. (2020a). 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]
- Grifoni A, Voic H, Dhanda SK, Kidd CK, Brien JD, Buus S, Stryhn A, Durbin AP, Whitehead S, Diehl SA, et al. (2020b). T Cell Responses Induced by Attenuated Flavivirus Vaccination Are Specific and Show Limited Cross-Reactivity with Other Flavivirus Species. J. Virol 94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grunst MW, and Uchil PD (2022). Fc effector cross-reactivity: A hidden arsenal against SARS-CoV-2’s evasive maneuvering. Cell Reports Medicine 3, 100540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guthmiller JJ, Han J, Utset HA, Li L, Lan LY-L, Henry C, Stamper CT, McMahon M, O’Dell G, Fernández-Quintero ML, et al. (2022). Broadly neutralizing antibodies target a haemagglutinin anchor epitope. Nature 602, 314–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Habel JR, Nguyen THO, van de Sandt CE, Juno JA, Chaurasia P, Wragg K, Koutsakos M, Hensen L, Jia X, Chua B, et al. (2020). Suboptimal SARS-CoV-2-specific CD8+ T cell response associated with the prominent HLA-A*02:01 phenotype. Proc. Natl. Acad. Sci. U. S. A 117, 24384–24391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hashiguchi T, Fusco ML, Bornholdt ZA, Lee JE, Flyak AI, Matsuoka R, Kohda D, Yanagi Y, Hammel M, Crowe JE Jr, et al. (2015). Structural basis for Marburg virus neutralization by a cross-reactive human antibody. Cell 160, 904–912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hastie KM, Cross RW, Harkins SS, Zandonatti MA, Koval AP, Heinrich ML, Rowland MM, Robinson JE, Geisbert TW, Garry RF, et al. (2019). Convergent Structures Illuminate Features for Germline Antibody Binding and Pan-Lassa Virus Neutralization. Cell 178, 1004–1015.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hastie KM, Li H, Bedinger D, Schendel SL, Dennison SM, Li K, Rayaprolu V, Yu X, Mann C, Zandonatti M, et al. (2021). Defining variant-resistant epitopes targeted by SARS-CoV-2 antibodies: A global consortium study. Science 374, 472–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hessell AJ, Hangartner L, Hunter M, Havenith CEG, Beurskens FJ, Bakker JM, Lanigan CMS, Landucci G, Forthal DN, Parren PWHI, et al. (2007). Fc receptor but not complement binding is important in antibody protection against HIV. Nature 449, 101–104. [DOI] [PubMed] [Google Scholar]
- Hsieh C-L, Werner AP, Leist SR, Stevens LJ, Falconer E, Goldsmith JA, Chou C-W, Abiona OM, West A, Westendorf K, et al. (2021). Stabilized coronavirus spike stem elicits a broadly protective antibody. Cell Rep. 37, 109929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Impagliazzo A, Milder F, Kuipers H, Wagner MV, Zhu X, Hoffman RMB, van Meersbergen R, Huizingh J, Wanningen P, Verspuij J, et al. (2015). A stable trimeric influenza hemagglutinin stem as a broadly protective immunogen. Science 349, 1301–1306. [DOI] [PubMed] [Google Scholar]
- Jendroszek A, and Kjaergaard M Nanoscale spatial dependence of avidity in an IgG1 antibody. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jennewein MF, MacCamy AJ, Akins NR, Feng J, Homad LJ, Hurlburt NK, Seydoux E, Wan Y-H, Stuart AB, Edara VV, et al. (2021). Isolation and characterization of cross-neutralizing coronavirus antibodies from COVID-19+ subjects. Cell Rep. 36, 109353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin J, Galaz-Montoya JG, Sherman MB, Sun SY, Goldsmith CS, O’Toole ET, Ackerman L, Carlson L-A, Weaver SC, Chiu W, et al. (2018). Neutralizing Antibodies Inhibit Chikungunya Virus Budding at the Plasma Membrane. Cell Host Microbe 24, 417–428.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jun S-R, Leuze MR, Nookaew I, Uberbacher EC, Land M, Zhang Q, Wanchai V, Chai J, Nielsen M, Trolle T, et al. (2015). Ebolavirus comparative genomics. FEMS Microbiol. Rev 39, 764–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalimuddin S, Tham CYL, Qui M, de Alwis R, Sim JXY, Lim JME, Tan H-C, Syenina A, Zhang SL, Le Bert N, et al. (2021). Early T cell and binding antibody responses are associated with COVID-19 RNA vaccine efficacy onset. Med (N Y) 2, 682–688.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keeton R, Richardson SI, Moyo-Gwete T, Hermanus T, Tincho MB, Benede N, Manamela NP, Baguma R, Makhado Z, Ngomti A, et al. (2021). Prior infection with SARS-CoV-2 boosts and broadens Ad26.COV2.S immunogenicity in a variant-dependent manner. Cell Host Microbe 29, 1611–1619.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keeton R, Tincho MB, Ngomti A, Baguma R, Benede N, Suzuki A, Khan K, Cele S, Bernstein M, Karim F, et al. (2022). T cell responses to SARS-CoV-2 spike cross-recognize Omicron. Nature. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keller MD, Harris KM, Jensen-Wachspress MA, Kankate VV, Lang H, Lazarski CA, Durkee-Shock J, Lee P-H, Chaudhry K, Webber K, et al. (2020). SARS-CoV-2-specific T cells are rapidly expanded for therapeutic use and target conserved regions of the membrane protein. Blood 136, 2905–2917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim AS, Kafai NM, Winkler ES, Gilliland TC Jr, Cottle EL, Earnest JT, Jethva PN, Kaplonek P, Shah AP, Fong RH, et al. (2021). Pan-protective anti-alphavirus human antibodies target a conserved E1 protein epitope. Cell 184, 4414–4429.e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- King LB, West BR, Moyer CL, Gilchuk P, Flyak A, Ilinykh PA, Bombardi R, Hui S, Huang K, Bukreyev A, et al. (2019). Cross-reactive neutralizing human survivor monoclonal antibody BDBV223 targets the ebolavirus stalk. Nat. Commun 10, 1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kotturi MF, Botten J, Sidney J, Bui H-H, Giancola L, Maybeno M, Babin J, Oseroff C, Pasquetto V, Greenbaum JA, et al. (2009). A multivalent and cross-protective vaccine strategy against arenaviruses associated with human disease. PLoS Pathog. 5, e1000695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kotturi MF, Botten J, Maybeno M, Sidney J, Glenn J, Bui H-H, Oseroff C, Crotty S, Peters B, Grey H, et al. (2010). Polyfunctional CD4+ T cell responses to a set of pathogenic arenaviruses provide broad population coverage. Immunome Res. 6, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kundu R, Narean JS, Wang L, Fenn J, Pillay T, Fernandez ND, Conibear E, Koycheva A, Davies M, Tolosa-Wright M, et al. (2022). Cross-reactive memory T cells associate with protection against SARS-CoV-2 infection in COVID-19 contacts. Nat. Commun 13, 80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LaBranche CC, McGuire AT, Gray MD, Behrens S, Kwong PD, Chen X, Zhou T, Sattentau QJ, Peacock J, Eaton A, et al. (2018). HIV-1 envelope glycan modifications that permit neutralization by germline-reverted VRC01-class broadly neutralizing antibodies. PLoS Pathog. 14, e1007431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Bert N, Tan AT, Kunasegaran K, Tham CYL, Hafezi M, Chia A, Chng MHY, Lin M, Tan N, Linster M, et al. (2020). SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 584, 457–462. [DOI] [PubMed] [Google Scholar]
- Le Bert N, Clapham HE, Tan AT, Chia WN, Tham CYL, Lim JM, Kunasegaran K, Tan LWL, Dutertre C-A, Shankar N, et al. (2021). Highly functional virus-specific cellular immune response in asymptomatic SARS-CoV-2 infection. J. Exp. Med 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee C-CD, Watanabe Y, Wu NC, Han J, Kumar S, Pholcharee T, Seabright GE, Allen JD, Lin C-W, Yang J-R, et al. (2021). A cross-neutralizing antibody between HIV-1 and influenza virus. PLoS Pathog. 17, e1009407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lehrer AT, Chuang E, Namekar M, Williams CA, Wong TAS, Lieberman MM, Granados A, Misamore J, Yalley-Ogunro J, Andersen H, et al. (2021). Recombinant Protein Filovirus Vaccines Protect Cynomolgus Macaques From Ebola, Sudan, and Marburg Viruses. Front. Immunol 12, 703986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W, Chen Y, Prévost J, Ullah I, Lu M, Gong SY, Tauzin A, Gasser R, Vézina D, Anand SP, et al. (2022). Structural basis and mode of action for two broadly neutralizing antibodies against SARS-CoV-2 emerging variants of concern. Cell Rep. 38, 110210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin Q, Li T, Chen Y, Lau S-Y, Wei M, Zhang Y, Zhang Z, Yao Q, Li J, Li Z, et al. (2018). Structural Basis for the Broad, Antibody-Mediated Neutralization of H5N1 Influenza Virus. J. Virol 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin W-S, Chen I-C, Chen H-C, Lee Y-C, and Wu S-C (2021). Glycan Masking of Epitopes in the NTD and RBD of the Spike Protein Elicits Broadly Neutralizing Antibodies Against SARS-CoV-2 Variants. Front. Immunol 12, 795741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lineburg KE, Grant EJ, Swaminathan S, Chatzileontiadou DSM, Szeto C, Sloane H, Panikkar A, Raju J, Crooks P, Rehan S, et al. (2021). CD8 T cells specific for an immunodominant SARS-CoV-2 nucleocapsid epitope cross-react with selective seasonal coronaviruses. Immunity 54, 1055–1065.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu J, Chandrashekar A, Sellers D, Barrett J, Jacob-Dolan C, Lifton M, McMahan K, Sciacca M, VanWyk H, Wu C, et al. (2022). Vaccines Elicit Highly Conserved Cellular Immunity to SARS-CoV-2 Omicron. Nature. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loyal L, Braun J, Henze L, Kruse B, Dingeldey M, Reimer U, Kern F, Schwarz T, Mangold M, Unger C, et al. (2021). Cross-reactive CD4+ T cells enhance SARS-CoV-2 immune responses upon infection and vaccination. Science 374, eabh1823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Madelon N, Heikkilä N, Royo IS, Fontannaz P, Breville G, Lauper K, Goldstein R, Grifoni A, Sette A, Siegrist C-A, et al. (2021). Omicron-specific cytotoxic T-cell responses are boosted following a third dose of mRNA COVID-19 vaccine in anti-CD20-treated multiple sclerosis patients. medRxiv 2021.12.20.21268128. [Google Scholar]
- Mallajosyula VVA, Citron M, Ferrara F, Lu X, Callahan C, Heidecker GJ, Sarma SP, Flynn JA, Temperton NJ, Liang X, et al. (2014). Influenza hemagglutinin stem-fragment immunogen elicits broadly neutralizing antibodies and confers heterologous protection. Proc. Natl. Acad. Sci. U. S. A 111, E2514–E2523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matassov D, Mire CE, Latham T, Geisbert JB, Xu R, Ota-Setlik A, Agans KN, Kobs DJ, Wendling MQS, Burnaugh A, et al. (2018). Single-Dose Trivalent VesiculoVax Vaccine Protects Macaques from Lethal Ebolavirus and Marburgvirus Challenge. J. Virol 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mateus J, Grifoni A, Tarke A, Sidney J, Ramirez SI, Dan JM, Burger ZC, Rawlings SA, Smith DM, Phillips E, et al. (2020). Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science 370, 89–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mateus J, Dan JM, Zhang Z, Rydyznski Moderbacher C, Lammers M, Goodwin B, Sette A, Crotty S, and Weiskopf D (2021). Low-dose mRNA-1273 COVID-19 vaccine generates durable memory enhanced by cross-reactive T cells. Science 374, eabj9853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melo-González F, Soto JA, González LA, Fernández J, Duarte LF, Schultz BM, Gálvez NMS, Pacheco GA, Ríos M, Vázquez Y, et al. (2021). Recognition of Variants of Concern by Antibodies and T Cells Induced by a SARS-CoV-2 Inactivated Vaccine. Front. Immunol 12, 747830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milligan JC, Parekh DV, Fuller KM, Igarashi M, Takada A, and Saphire EO (2019). Structural Characterization of Pan-Ebolavirus Antibody 6D6 Targeting the Fusion Peptide of the Surface Glycoprotein. J. Infect. Dis 219, 415–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milligan JC, Davis CW, Yu X, Ilinykh PA, Huang K, Halfmann PJ, Cross RW, Borisevich V, Agans KN, Geisbert JB, et al. (2022). Asymmetric and non-stoichiometric glycoprotein recognition by two distinct antibodies results in broad protection against ebolaviruses. Cell 185, 995–1007.e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milligan JC, Davis CW, Yu X, Ilinykh PA, Huang K, Halfmann P, Cross RW, Borisevich V, Agans KN, Geisbert JB, Chennareddy C, Goff AJ, Piper AE, Hui S, Shaffer K, Buck T, Heinrich ML, Branco LM, Crozier I, Holbrook MR, Kuhn JH, Kawaoka Y, Glass PJ, Bukreyev A, Geisbert TW, Worwa G, Ahmed R, and Saphire EO (2022). Asymmetric and non-stoichiometric glycoprotein recognition by two distinct antibodies results in broad protection against ebolaviruses. Cell. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neerukonda SN, Vassell R, Herrup R, Liu S, Wang T, Takeda K, Yang Y, Lin T-L, Wang W, and Weiss CD (2021). Establishment of a well-characterized SARS-CoV-2 lentiviral pseudovirus neutralization assay using 293T cells with stable expression of ACE2 and TMPRSS2. PLoS One 16, e0248348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nelde A, Bilich T, Heitmann JS, Maringer Y, Salih HR, Roerden M, Lübke M, Bauer J, Rieth J, Wacker M, et al. (2021). SARS-CoV-2-derived peptides define heterologous and COVID-19-induced T cell recognition. Nat. Immunol 22, 74–85. [DOI] [PubMed] [Google Scholar]
- Ng KW, Faulkner N, Wrobel AG, Gamblin SJ, and Kassiotis G (2021). Heterologous humoral immunity to human and zoonotic coronaviruses: Aiming for the achilles heel. Semin. Immunol 55, 101507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nguyen DN, Xu B, Stanfield RL, Bailey JK, Horiya S, Temme JS, Leon DR, LaBranche CC, Montefiori DC, Costello CE, et al. (2019). Oligomannose Glycopeptide Conjugates Elicit Antibodies Targeting the Glycan Core Rather than Its Extremities. ACS Cent Sci 5, 237–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oda M, and Azuma T (2000). Reevaluation of stoichiometry and affinity/avidity in interactions between anti-hapten antibodies and mono- or multi-valent antigens. Mol. Immunol 37, 1111–1122. [DOI] [PubMed] [Google Scholar]
- Peng Y, Felce SL, Dong D, Penkava F, Mentzer AJ, Yao X, Liu G, Yin Z, Chen J-L, Lu Y, et al. (2021). An immunodominant NP105–113-B*07:02 cytotoxic T cell response controls viral replication and is associated with less severe COVID-19 disease. Nat. Immunol 23, 50–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pinto D, Fenwick C, Caillat C, Silacci C, Guseva S, Dehez F, Chipot C, Barbieri S, Minola A, Jarrossay D, et al. (2019). Structural Basis for Broad HIV-1 Neutralization by the MPER-Specific Human Broadly Neutralizing Antibody LN01. Cell Host Microbe 26, 623–637.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pinto D, Sauer MM, Czudnochowski N, Low JS, Tortorici MA, Housley MP, Noack J, Walls AC, Bowen JE, Guarino B, et al. (2021). Broad betacoronavirus neutralization by a stem helix-specific human antibody. Science 373, 1109–1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prakash S, Srivastava R, Coulon P-G, Dhanushkodi NR, Chentoufi AA, Tifrea DF, Edwards RA, Figueroa CJ, Schubl SD, Hsieh L, et al. (2021). Genome-Wide B Cell, CD4, and CD8 T Cell Epitopes That Are Highly Conserved between Human and Animal Coronaviruses, Identified from SARS-CoV-2 as Targets for Preemptive Pan-Coronavirus Vaccines. J. Immunol 206, 2566–2582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Redd AD, Nardin A, Kared H, Bloch EM, Pekosz A, Laeyendecker O, Abel B, Fehlings M, Quinn TC, and Tobian AAR (2021). CD8+ T-Cell Responses in COVID-19 Convalescent Individuals Target Conserved Epitopes From Multiple Prominent SARS-CoV-2 Circulating Variants. Open Forum Infect Dis 8, ofab143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Regla-Nava JA, Elong Ngono A, Viramontes KM, Huynh A-T, Wang Y-T, Nguyen A-VT, Salgado R, Mamidi A, Kim K, Diamond MS, et al. (2018). Cross-reactive Dengue virus-specific CD8+ T cells protect against Zika virus during pregnancy. Nat. Commun 9, 3042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Remmel JL, Frei JC, Butler SE, Lai JR, and Ackerman ME (2021). Diverse contributions of avidity to the broad neutralization of Dengue virus by antibodies targeting the E dimer epitope. Virology 559, 57–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roozendaal R, Hendriks J, van Effelterre T, Spiessens B, Dekking L, Solforosi L, Czapska-Casey D, Bockstal V, Stoop J, Splinter D, et al. (2020). Nonhuman primate to human immunobridging to infer the protective effect of an Ebola virus vaccine candidate. NPJ Vaccines 5, 112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rydyznski Moderbacher C, Ramirez SI, Dan JM, Grifoni A, Hastie KM, Weiskopf D, Belanger S, Abbott RK, Kim C, Choi J, et al. (2020). Antigen-Specific Adaptive Immunity to SARS-CoV-2 in Acute COVID-19 and Associations with Age and Disease Severity. Cell 183, 996–1012.e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sabatino JJ Jr, Mittl K, Rowles WM, McPolin K, Rajan JV, Laurie MT, Zamecnik CR, Dandekar R, Alvarenga BD, Loudermilk RP, et al. (2022). Multiple sclerosis therapies differentially affect SARS-CoV-2 vaccine-induced antibody and T cell immunity and function. JCI Insight 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sagar M, Reifler K, Rossi M, Miller NS, Sinha P, White LF, and Mizgerd JP (2021). Recent endemic coronavirus infection is associated with less-severe COVID-19. J. Clin. Invest 131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sauer MM, Tortorici MA, Park Y-J, Walls AC, Homad L, Acton OJ, Bowen JE, Wang C, Xiong X, de van der Schueren W, et al. (2021). Structural basis for broad coronavirus neutralization. Nat. Struct. Mol. Biol 28, 478–486. [DOI] [PubMed] [Google Scholar]
- Saunders KO, Nicely NI, Wiehe K, Bonsignori M, Meyerhoff RR, Parks R, Walkowicz WE, Aussedat B, Wu NR, Cai F, et al. (2017). Vaccine Elicitation of High Mannose-Dependent Neutralizing Antibodies against the V3-Glycan Broadly Neutralizing Epitope in Nonhuman Primates. Cell Rep. 18, 2175–2188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schouest B, Grifoni A, Pham J, Mateus J, Sydney J, Brien JD, De Silva AD, Balmaseda A, Harris E, Sette A, et al. (2021). Pre-existing T Cell Memory against Zika Virus. J. Virol 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schulien I, Kemming J, Oberhardt V, Wild K, Seidel LM, Killmer S, Sagar, Daul F, Salvat Lago M, Decker A, et al. (2021). Characterization of pre-existing and induced SARS-CoV-2-specific CD8 T cells. Nat. Med 27, 78–85. [DOI] [PubMed] [Google Scholar]
- Sekine T, Perez-Potti A, Rivera-Ballesteros O, Strålin K, Gorin J-B, Olsson A, Llewellyn-Lacey S, Kamal H, Bogdanovic G, Muschiol S, et al. (2020). Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild COVID-19. Cell 183, 158–168.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sette A, and Crotty S (2020). Pre-existing immunity to SARS-CoV-2: the knowns and unknowns. Nat. Rev. Immunol 20, 457–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shomuradova AS, Vagida MS, Sheetikov SA, Zornikova KV, Kiryukhin D, Titov A, Peshkova IO, Khmelevskaya A, Dianov DV, Malasheva M, et al. (2020). SARS-CoV-2 Epitopes Are Recognized by a Public and Diverse Repertoire of Human T Cell Receptors. Immunity 53, 1245–1257.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sidney J, Peters B, and Sette A (2020). Epitope prediction and identification-adaptive T cell responses in humans. Semin. Immunol 50, 101418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- da Silva Antunes R, Pallikkuth S, Williams E, Dawen Yu E, Mateus J, Quiambao L, Wang E, Rawlings SA, Stadlbauer D, Jiang K, et al. (2021). Differential T-Cell Reactivity to Endemic Coronaviruses and SARS-CoV-2 in Community and Health Care Workers. J. Infect. Dis 224, 70–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soresina A, Moratto D, Chiarini M, Paolillo C, Baresi G, Focà E, Bezzi M, Baronio B, Giacomelli M, and Badolato R (2020). Two X-linked agammaglobulinemia patients develop pneumonia as COVID-19 manifestation but recover. Pediatr. Allergy Immunol 31, 565–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swadling L, Diniz MO, Schmidt NM, Amin OE, Chandran A, Shaw E, Pade C, Gibbons JM, Le Bert N, Tan AT, et al. (2022). Pre-existing polymerase-specific T cells expand in abortive seronegative SARS-CoV-2. Nature 601, 110–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan AT, Linster M, Tan CW, Le Bert N, Chia WN, Kunasegaran K, Zhuang Y, Tham CYL, Chia A, Smith GJD, et al. (2021). Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients. Cell Rep. 34, 108728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tarke A, Sidney J, Methot N, Yu ED, Zhang Y, Dan JM, Goodwin B, Rubiro P, Sutherland A, Wang E, et al. (2021a). Impact of SARS-CoV-2 variants on the total CD4 and CD8 T cell reactivity in infected or vaccinated individuals. Cell Rep Med 2, 100355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tarke A, Coelho CH, Zhang Z, Dan JM, Yu ED, Methot N, Bloom NI, Goodwin B, Phillips E, Mallal S, et al. (2021b). SARS-CoV-2 vaccination induces immunological memory able to cross-recognize variants from Alpha to Omicron. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tarke A, Coelho CH, Zhang Z, Dan JM, Yu ED, Methot N, Bloom NI, Goodwin B, Phillips E, Mallal S, et al. (2022). SARS-CoV-2 vaccination induces immunological T cell memory able to cross-recognize variants from Alpha to Omicron. Cell. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terajima M, Cruz J, Co MDT, Lee J-H, Kaur K, Wrammert J, Wilson PC, and Ennis FA (2011). Complement-dependent lysis of influenza a virus-infected cells by broadly cross-reactive human monoclonal antibodies. J. Virol 85, 13463–13467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thulin NK, and Wang TT (2018). The Role of Fc Gamma Receptors in Broad Protection against Influenza Viruses. Vaccines (Basel) 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai W-Y, Lai C-Y, Wu Y-C, Lin H-E, Edwards C, Jumnainsong A, Kliks S, Halstead S, Mongkolsapaya J, Screaton GR, et al. (2013). High-avidity and potently neutralizing cross-reactive human monoclonal antibodies derived from secondary dengue virus infection. J. Virol 87, 12562–12575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai W-Y, Chen H-L, Tsai J-J, Dejnirattisai W, Jumnainsong A, Mongkolsapaya J, Screaton G, Crowe JE Jr, and Wang W-K (2018). Potent Neutralizing Human Monoclonal Antibodies Preferentially Target Mature Dengue Virus Particles: Implication for Novel Strategy for Dengue Vaccine. J. Virol 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tyagi A, Ahmed T, Shi J, and Bhushan S (2020). A complex between the Zika virion and the Fab of a broadly cross-reactive neutralizing monoclonal antibody revealed by cryo-EM and single particle analysis at 4.1 Å resolution. J Struct Biol X 4, 100028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- VanBlargan LA, Goo L, and Pierson TC (2016). Deconstructing the Antiviral Neutralizing-Antibody Response: Implications for Vaccine Development and Immunity. Microbiol. Mol. Biol. Rev 80, 989–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vanderheijden N, Stevaert A, Xie J, Ren X, Barbezange C, Noppen S, Desombere I, Verhasselt B, Geldhof P, Vereecke N, et al. (2021). Functional Analysis of Human and Feline Coronavirus Cross-Reactive Antibodies Directed Against the SARS-CoV-2 Fusion Peptide. Front. Immunol 12, 790415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walls AC, Miranda MC, Schäfer A, Pham MN, Greaney A, Arunachalam PS, Navarro M-J, Tortorici MA, Rogers K, O’Connor MA, et al. (2021). Elicitation of broadly protective sarbecovirus immunity by receptor-binding domain nanoparticle vaccines. Cell 184, 5432–5447.e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C, van Haperen R, Gutiérrez-Álvarez J, Li W, Okba NMA, Albulescu I, Widjaja I, van Dieren B, Fernandez-Delgado R, Sola I, et al. (2021a). A conserved immunogenic and vulnerable site on the coronavirus spike protein delineated by cross-reactive monoclonal antibodies. Nat. Commun 12, 1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang P, Nair MS, Liu L, Iketani S, Luo Y, Guo Y, Wang M, Yu J, Zhang B, Kwong PD, et al. (2021b). Antibody Resistance of SARS-CoV-2 Variants B.1.351 and B.1.1.7. bioRxiv. [DOI] [PubMed] [Google Scholar]
- Weiskopf D, Angelo MA, de Azeredo EL, Sidney J, Greenbaum JA, Fernando AN, Broadwater A, Kolla RV, De Silva AD, de Silva AM, et al. (2013). Comprehensive analysis of dengue virus-specific responses supports an HLA-linked protective role for CD8+ T cells. Proc. Natl. Acad. Sci. U. S. A 110, E2046–E2053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weiskopf D, Angelo MA, Sidney J, Peters B, Shresta S, and Sette A (2014). Immunodominance changes as a function of the infecting dengue virus serotype and primary versus secondary infection. J. Virol 88, 11383–11394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weiskopf D, Angelo MA, Bangs DJ, Sidney J, Paul S, Peters B, de Silva AD, Lindow JC, Diehl SA, Whitehead S, et al. (2015). The human CD8+ T cell responses induced by a live attenuated tetravalent dengue vaccine are directed against highly conserved epitopes. J. Virol 89, 120–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weiskopf D, Angelo MA, Grifoni A, O’Rourke PH, Sidney J, Paul S, De Silva AD, Phillips E, Mallal S, Premawansa S, et al. (2016). HLA-DRB1 Alleles Are Associated With Different Magnitudes of Dengue Virus-Specific CD4+ T-Cell Responses. J. Infect. Dis 214, 1117–1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wen J, Tang WW, Sheets N, Ellison J, Sette A, Kim K, and Shresta S (2017). Identification of Zika virus epitopes reveals immunodominant and protective roles for dengue virus cross-reactive CD8+ T cells. Nat Microbiol 2, 17036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wen J, Wang Y-T, Valentine KM, Dos Santos Alves RP, Xu Z, Regla-Nava JA, Ngono AE, Young MP, Ferreira LCS, and Shresta S (2020). CD4+ T Cells Cross-Reactive with Dengue and Zika Viruses Protect against Zika Virus Infection. Cell Rep. 31, 107566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- West BR, Moyer CL, King LB, Fusco ML, Milligan JC, Hui S, and Saphire EO (2018). Structural Basis of Pan-Ebolavirus Neutralization by a Human Antibody against a Conserved, yet Cryptic Epitope. MBio 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whittle JRR, Zhang R, Khurana S, King LR, Manischewitz J, Golding H, Dormitzer PR, Haynes BF, Walter EB, Moody MA, et al. (2011). Broadly neutralizing human antibody that recognizes the receptor-binding pocket of influenza virus hemagglutinin. Proc. Natl. Acad. Sci. U. S. A 108, 14216–14221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams WB, Meyerhoff RR, Edwards RJ, Li H, Manne K, Nicely NI, Henderson R, Zhou Y, Janowska K, Mansouri K, et al. (2021). Fab-dimerized glycan-reactive antibodies are a structural category of natural antibodies. Cell 184, 2955–2972.e25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williamson LE, Reeder KM, Bailey K, Tran MH, Roy V, Fouch ME, Kose N, Trivette A, Nargi RS, Winkler ES, et al. (2021). Therapeutic alphavirus cross-reactive E1 human antibodies inhibit viral egress. Cell 184, 4430–4446.e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu NC, and Wilson IA (2020). Influenza Hemagglutinin Structures and Antibody Recognition. Cold Spring Harb. Perspect. Med 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamayoshi S, Yasuhara A, Ito M, Akasaka O, Nakamura M, Nakachi I, Koga M, Mitamura K, Yagi K, Maeda K, et al. (2021). Antibody titers against SARS-CoV-2 decline, but do not disappear for several months. EClinicalMedicine 32, 100734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang F, Mariz FC, Zhao X, Spagnoli G, Ottonello S, and Müller M (2020). Broad Neutralization Responses Against Oncogenic Human Papillomaviruses Induced by a Minor Capsid L2 Polytope Genetically Incorporated Into Bacterial Ferritin Nanoparticles. Front. Immunol 11, 606569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yauch LE, Zellweger RM, Kotturi MF, Qutubuddin A, Sidney J, Peters B, Prestwood TR, Sette A, and Shresta S (2009). A protective role for dengue virus-specific CD8+ T cells. J. Immunol 182, 4865–4873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yauch LE, Prestwood TR, May MM, Morar MM, Zellweger RM, Peters B, Sette A, and Shresta S (2010). CD4+ T cells are not required for the induction of dengue virus-specific CD8+ T cell or antibody responses but contribute to protection after vaccination. J. Immunol 185, 5405–5416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin V, Lai S-H, Caniels TG, Brouwer PJM, Brinkkemper M, Aldon Y, Liu H, Yuan M, Wilson IA, Sanders RW, et al. Probing Affinity, Avidity, Anti-Cooperativity, and Competition in Antibody and Receptor Binding to the SARS-CoV-2 Spike by Single Particle Mass Analyses. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zellweger RM, Tang WW, Eddy WE, King K, Sanchez MC, and Shresta S (2015). CD8+ T Cells Can Mediate Short-Term Protection against Heterotypic Dengue Virus Reinfection in Mice. J. Virol 89, 6494–6505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou P, Yuan M, Song G, Beutler N, Shaabani N, Huang D, He W-T, Zhu X, Callaghan S, Yong P, et al. (2022). A human antibody reveals a conserved site on beta-coronavirus spike proteins and confers protection against SARS-CoV-2 infection. bioRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]

