ABSTRACT
The COVID-19 pandemic has greatly enhanced our understanding of CD8+ T cell immunity and their role in natural infection and vaccine-induced protection. Rapid and early SARS-CoV-2-specific CD8+ T cell responses have been associated with efficient viral clearance and mild disease. Virus-specific CD8+ T cell responses can compensate for waning, morbidity-related, and iatrogenic reduction of humoral immunity. After infection or vaccination, SARS-CoV-2-specific memory CD8+ T cells are formed, which mount an efficient recall response in the event of breakthrough infection and help to protect from severe disease. Due to their breadth and ability to target mainly highly conserved epitopes, SARS-CoV-2-specific CD8+ T cells are also able to cross-recognize epitopes of viral variants, thus maintaining immunity even after the emergence of viral evolution. In some cases, however, CD8+ T cells may contribute to the pathogenesis of severe COVID-19. In particular, delayed and uncontrolled, e.g., nonspecific and hyperactivated, cytotoxic CD8+ T cell responses have been linked to poor COVID-19 outcomes. In this minireview, we summarize the tremendous knowledge about CD8+ T cell responses to SARS-CoV-2 infection and COVID-19 vaccination that has been gained over the past 5 years, while also highlighting the critical knowledge gaps that remain.
KEYWORDS: CD8+ T cells, SARS-CoV-2, COVID-19, virus-specific immunity
INTRODUCTION
The SARS-CoV-2 pandemic has taught important lessons about the central role of virus-specific CD8+ T cells in mediating antiviral immunity, both during natural infection and following vaccination. In contrast to antibodies, which primarily prevent viral entry, CD8+ T cells exert antiviral functions by recognizing viral peptides presented on infected cells, allowing them to directly kill the virus-infected cells and prevent further viral spread. This is particularly important for individuals with reduced or absent humoral responses. Indeed, virus-specific CD8+ T cell responses induced by infection are associated with improved survival and thus, with a protective role, in immunosuppressed individuals, who have deficient humoral and B cell immunity (1). The protective role of CD8+ T cells during viral infection has further been demonstrated in preclinical models where depletion of CD8+ T cells abrogated viral control and CD8+ T cell transfer improved viral control (2). Moreover, CD8+ T cells have been suggested to play a significant role in the control of emerging viral variants. CD8+ T cells target a broad range of epitopes from both structural and nonstructural proteins of the virus, which are often conserved across different strains (3–5). The breadth of the CD8+ T cell response together with the high level of conservation enables virus-specific CD8+ T cells to cross-recognize various viral variants (4–7), thereby mitigating the impact of mutations that may arise over time. Consequently, a robust CD8+ T cell response can be crucial in maintaining immunity and preventing severe disease, even in the face of viral evolution. Infection- and vaccine-induced CD8+ T cell responses are stable over time and are rapidly reactivated in case of reinfection, hence exhibiting robust memory responses that contribute to long-lasting immunity (8, 9). However, despite these beneficial effects, CD8+ T cells have also been implicated in the pathogenesis of severe COVID-19. They may be involved indirectly by an insufficient ability to limit viral replication and spread or directly through nonspecific effector functions that lead to the killing of uninfected cells and tissue damage. In this minireview, we highlight some of the key features of SARS-CoV-2-specific CD8+ T cell immunity during natural infection and after vaccination that contribute to both viral control and disease pathogenesis.
ROLE OF VIRUS-SPECIFIC CD8+ T CELLS DURING SARS-COV-2 INFECTION
The COVID-19 pandemic and the tremendous efforts to understand the correlates of T-cell-mediated immune protection have yielded immense novel insights into the role of virus-specific CD8+ T cells during an acute human viral infection. In fact, by using several different methodological approaches ranging from functional and phenotypic to transcriptomic analyses, multiple groups have investigated the basic principles of SARS-CoV-2-specific CD8+ T cell immunity during acute and resolved infection. The key findings can be summarized as follows:
First, virus-specific CD8+ T cells are rapidly induced in approximately 70%–90% of COVID-19 cases within 1 month after infection and display an estimated half-life of ~200 days (10–13). By longitudinal analyses, we identified the peak of virus-specific CD8+ T cells as early as 1 week postinfection (12). At these early time points, the SARS-CoV-2-specific CD8+ T cells highly express activation markers such as CD38, CD69, or PD-1 (Fig. 1). The expression of PD-1 is thereby linked to activation and not exhaustion as these cells show no functional impairments (14). In fact, SARS-CoV-2-specific CD8+ T cells display high expression levels of effector molecules including IFN-γ, TNF, and CD107a as a surrogate for degranulation. This polyfunctional profile was present during the expansion phase (~30 days) as well as at later time points (>180 days after symptom onset) (10, 12). After the expansion phase, the virus-specific CD8+ T cell response shows a prolonged contraction phase and subsequently displays an effector memory differentiation state, as indicated by the expressions of CD45RA, CX3CR1, KLRG1, and CD57 (12) (Fig. 1). Although some heterogeneity has been reported in the phenotype and function of SARS-CoV-2-specific CD8+ T cells during acute viral infection, the majority of studies suggest that the SARS-CoV-2-specific CD8+ T cell immunity follows the classical T cell differentiation program described for other acute self-limiting infections. Notably, the SARS-CoV-2-specific CD8+ T cell response is multispecific and targets an estimated median of 17 targeted epitopes per individual with an immunodominance toward epitopes derived from nucleo- and membrane proteins (15) (Fig. 2).
Fig 1.

Kinetics of SARS-CoV-2-specific CD8+ T cell responses after natural infection. Early after infection, virus-specific CD8+ T cells expand, while forming a heterogeneous pool of highly activated effector CD8+ T cells and memory precursors. After viral clearance, the virus-specific CD8+ T cell response contracts, resulting in a stable pool of memory CD8+ T cells that persist long term.
Fig 2.

SARS-CoV-2-specific CD8+ T cell responses in lung and peripheral blood. Natural infection induces expansion of multispecific CD8+ T cells in the lung and peripheral blood, whereas (mRNA) vaccination primarily induces spike-specific CD8+ T cells in the periphery. After peak response, virus-specific memory CD8+ T cells are formed, which persist in the lungs and blood.
Second, SARS-CoV-2-specific CD8+ T cells are readily detectable in the upper respiratory tract and lung tissue of infected patients, even at higher frequencies when compared to peripheral blood (16) (Fig. 2). These tissue-resident SARS-CoV-2-specific CD8+ T cells with high functionality are even present in cases where circulating virus-specific CD8+ T cells are barely detectable (17), indicating a specific enrichment at the site of the disease. Tissue-resident CD8+ T cells found in the airways of SARS-CoV-2-infected patients, who survived COVID-19, exhibited an activated and functional profile indicating their ability to kill virus-infected cells and mediate viral clearance. In line with this, a larger proportion of effector CD8+ T cells with characteristics of tissue residency were found in bronchoalveolar lavage fluid obtained from SARS-CoV-2-infected subjects with moderate compared to severe COVID-19 (18, 19). The virus-specific CD8+ T cells also appear to be long-lived at the site of infection as virus-specific memory CD8+ T cells are still detectable in the lungs, lung-associated lymph nodes, and nasal mucosa for up to 12 months after the last antigen exposure (17, 20–22) (Fig. 2). Nevertheless, it is important to note that the dynamics of emergence and persistence of virus-specific CD8+ T cells differ between anatomical sites as they are colonized by distinct T cell populations with different abilities to persist. For instance, the nasal turbinates exhibit exclusively CD8+ T cells, while the nasopharyngeal adenoids are characterized by CD4+ and CD8+ T cells (23). Moreover, the persistence of memory T cells may also differ between the upper respiratory tract and the lungs. In fact, SARS-CoV-2-specific T cells appear to reduce in the nasal fluid 3–4 months after infection (24), whereas they are detectable for more than 1 year in the nasal tissue (22) and for up to 10 months in the lungs (17).
Third, SARS-CoV-2 infection leads to the formation of memory precursor CD8+ T cells that are characterized, e.g., by the expression of classical differentiation, transcription, and pro-survival markers such as CD45RA, CD127, TCF-1, and BCL-2 (11, 12) (Fig. 1). These cells are relevant for maintaining the CD8+ T cell response and for the generation of a robust, long-lasting CD8+ T cell memory pool. Within the pool of circulating SARS-CoV-2-specific CD8+ T memory cells, different memory subsets exist, such as central memory (TCM), effector memory (TEM), and terminally differentiated effector memory expressing CD45RA (TEMRA) T cells (12). Among these different CD8+ T cell memory subsets, TEMRA cells have been reported to be the most abundant subset (11, 25). These cells have also been associated with protection against several other viral pathogens, including influenza virus (IV), cytomegalovirus (CMV), Epstein–Barr virus (EBV), and human immunodeficiency virus (HIV) (26–29). The virus-specific CD8+ T cell memory remains stable over time and is still present long after infection (30) (Fig. 1). These cells are characterized by a transcriptional signature that marks long-lived, circulating human memory CD8+ T cells following an acute viral infection (31). SARS-CoV-2-specific memory CD8+ T cells are fully functional and similar to other viral infections, such as influenza virus (12), even though they are present at a slightly lower frequency.
Lastly, and most importantly, early SARS-CoV-2-specific CD8+ T cell responses are associated with a reduced disease severity and an asymptomatic course of primary infection (32–34). In fact, the emergence of a rapid, robust, and polyfunctional CD8+ T cell response in the periphery has been shown to correlate with lower peak viral loads, rapid viral clearance, and a better overall clinical outcome (35). One study even observed a higher cytokine production per T cell in asymptomatic infection, affirming their high functionality and antiviral efficacy (36). The critical role of virus-specific CD8+ T cells in mediating antiviral immunity has also been demonstrated by their presence in individuals exposed to SARS-CoV-2 who did not become infected, suggesting that early T cell responses can facilitate rapid viral clearance before seroconversion (34, 37, 38). Conversely, more severe cases of COVID-19 have been associated with a delayed induction and a lower frequency of SARS-CoV-2-specific CD8+ T cells in the peripheral blood (39, 40) and lung tissue (19, 41), as well as a disproportional secretion of inflammatory cytokines (36). Hence, the emergence of an early and functionally competent virus-specific CD8+ T cell response seems critical for a mild course of disease. This conclusion is further supported by the finding that protective CD8+ T cell immunity is present in immunosuppressed patients with reduced humoral responses and B cell deficiency. CD8+ T cell responses are also associated with an improved survival of SARS-CoV-2-infected patients with hematologic cancers who have impaired humoral responses and are treated with anti-CD20 therapy (1). Noteworthily, unlike antibody-mediated protection, CD8+ T cells maintain their protective effects regardless of which epitope of the viral proteins they target or where these epitopes are located within the virus. Furthermore, the protective efficacy of CD8+ T cells in SARS-CoV-2 infection has been shown in macaques, where depletion of CD8+ T cells after SARS-CoV-2 infection caused increased viral loads following re-challenge with SARS-CoV-2 (42).
CD8+ T CELL RESPONSES AFTER VACCINATION
The development of efficient COVID-19 vaccines and subsequent international vaccination campaigns have successfully reduced the risk of primary SARS-CoV-2 infection and severe disease. Many different vaccine platforms have been tested in humans, and several of them have been approved. The most widely used vaccines are from Pfizer-BioNTech (Comirnaty), Oxford-AstraZeneca (Vaxzevria), Moderna (Spikevax), Johnson & Johnson (Jcovden), Sinopharm BIBP (Covilo), Sinovac (CoronaVac), and Novavax (Nuvaxovid) and represent mRNA-, adenoviral vector-, inactivated virus-, and protein-based vaccine technologies (https://covid19.trackvaccines.org/agency/who/). The different vaccine platforms elicit distinct T cell responses. For example, the inactivated virus vaccine induces robust CD4+ T cell responses in most individuals, while CD8+ T cell responses are hardly detectable (43). Still, a key advantage of this vaccine is its ability to induce T cell responses not only against the spike protein but also against all other viral proteins, many of which tend to be more conserved across different viral variants (44, 45). In contrast, mRNA and adenoviral vector vaccines that are used in most countries (https://covid19.trackvaccines.org/agency/who/) are solely based on the spike protein and, therefore, elicit only spike-specific T cell responses (Fig. 2). These responses, however, are robust and mediated by both CD4+ and CD8+ T cells, in most individuals (46, 47).
Early studies on SARS-CoV-2-specific CD8+ T cells following vaccination with mRNA- and adenoviral vector-based regimens have demonstrated a rapid induction of spike-specific CD8+ T cells in most individuals as early as 5–7 days after the initial (prime) immunization (Fig. 2). Indeed, by performing a longitudinal analysis starting at the baseline of prime vaccination until several months after boost, we observed in agreement with other reports (48) a rapid and stable mobilization of spike-specific CD8+ T cells by SARS-CoV-2 mRNA vaccination at a time point when neutralizing antibodies were hardly detectable (9). The peak mobilization of neutralizing antibodies is first detectable after boost, most likely reflecting the maturation of this response in the secondary lymphoid organs before they are released to the circulation. A strong induction of spike-specific CD8+ T cells following mRNA vaccination has also been observed in immunosuppressed patients, such as those receiving anti-CD20 therapy and liver transplant recipients, despite their impaired humoral responses (49–52). Although the frequencies of the vaccine-induced CD8+ T cell responses may be lower in immunosuppressed compared to immunocompetent individuals, they are still functionally competent in most individuals (51, 53). Indeed, the functional capacity of spike-specific CD8+ T cells after mRNA vaccination is comparable to those induced by natural infection, albeit slight differences in the phenotype and kinetics. For example, a lower expression of the activation marker CD38 has been observed on early memory spike-specific CD8+ T cells after vaccination compared to natural infection (9, 12), indicating enhanced activation after infection with different kinetics. In line with this, a steeper contraction of the spike-specific CD8+ T cell response has been reported after vaccination compared to the prolonged contraction phase detected after natural infection (9, 12). Most likely, the differential route and duration of antigen contact contribute to these differences.
Importantly, as in natural infection, vaccination alone is also capable of inducing tissue-resident SARS-CoV-2-specific CD8+ T cells in nasal mucosal tissues, where they can rapidly respond to subsequent infection (22). In line with this, the first appearance of vaccine-induced SARS-CoV-2-specific CD8+ T cells coincides with the first protective clinical effect that can be observed within 12 days after the first vaccination, clearly supporting a potential protective role of virus-specific CD8+ T cell immunity (54). This is also supported by studies performed in nonhuman primates. Indeed, vaccinated macaques with strong virus-specific CD8+ T cell responses are able to control infection in the absence of SARS-CoV-2-specific neutralizing antibodies (55). The important role of vaccine-induced virus-specific CD8+ T cells in viral clearance is further supported by a study in vaccinated macaques where depletion of CD8+ T cells prior to infectious challenge prolonged viral infection (2). Moreover, vaccine failures in experimental Omicron challenge studies were linked to a lack of CD8+ T cell immunity, despite the presence of moderate antibody titers (56), further emphasizing the critical protective role of CD8+ T cells in viral infections.
The virus-specific CD8+ T cell response can be further enhanced by a booster vaccine. However, variations in the frequency of virus-specific CD8+ T cells have been observed depending on whether a homologous or heterologous booster vaccine was administered. Individuals who received an mRNA vaccine as a prime vaccination and a boost with the adenoviral vector-based vaccine Jcovden (heterologous booster) elicited higher spike-specific CD8+ T cell responses compared to those who received homologous booster (57). Similarly, individuals who received a prime vaccination with an adenoviral vector-based vaccine (Vaxzevria) and were boosted with an mRNA vaccine exhibited stronger spike-specific CD8+ T cell responses compared to individuals who received a homologous prime/boost vaccine regimen (58). Still, the overall effectiveness in protecting from severe disease is comparable between heterologous versus homologous regimens (59). In-depth studies of mRNA-vaccinated healthy individuals demonstrated that boosting with a second, third, or fourth dose of the mRNA vaccine results each time in a robust expansion of highly activated spike-specific effector CD8+ T cells that peak around 5–6 days after the respective boosting. This rapid expansion in spike-specific CD8+ T cells lasts approximately 30–60 days and is followed by a subsequent contraction phase, in which CD8+ T cell frequencies decrease until they reach frequencies similar to those observed before the boost vaccination (8, 9). Importantly, spike-specific CD8+ T cells retain their functional capacity throughout booster vaccination. This has been shown for both healthy and immunosuppressed individuals (8, 9, 51). Overall, these findings underline that vaccination induces fully functional CD8+ T cells with robust recall responses.
Vaccination leads to the stable formation of CD8+ T cell memory, as has been reported for natural infection (Fig. 2). However, the distribution of the different memory subsets varies slightly. For example, while higher numbers of early differentiated memory subsets, such as early differentiated, central memory, and transitional memory T cells, emerge after natural infection, higher frequencies of effector memory CD8+ T cells are present after vaccination (9). Furthermore, mRNA vaccination and natural infection induce the formation of stem cell-like memory CD8+ T cells (TSCM). These cells have previously been identified in individuals who received the yellow fever virus vaccine, which is highly effective and induces long-term immunity in humans (60). TSCM have self-renewal capacities and are, therefore, essential for the generation of long-lasting CD8+ T cell immunity. Spike-specific CD8+ TSCM cells are already detectable after prime vaccination (8, 61) and remain stable in frequency and phenotype over time and throughout further booster vaccinations (8). This indicates that vaccination induces the formation of a long-lived CD8+ T cell memory pool. Stable CD8+ T cell memory formation has been observed for at least 6–8 months after both vaccination regimens, adenoviral vector-based vaccination (62) and mRNA vaccination (63, 64). Yet, differences in the frequencies of memory CD8+ T cells may occur in individual cases. For example, reduced long-term cellular responses have been observed in high-risk populations such as elderly people and immunosuppressed individuals (51, 65, 66). However, although the spike-specific CD8+ T cell frequencies are reduced, they appear to remain stable over time (51), suggesting long-term persistence even in high-risk populations.
BREAKTHROUGH INFECTION AND HYBRID IMMUNITY
Virus-specific CD8+ T cells also play an important role in the context of breakthrough infection that is defined as an infection occurring in individuals despite prior vaccination against the same pathogen. Although vaccine-induced virus-specific CD8+ T cells do not necessarily protect vaccinated individuals from infection (67, 68), they still may play a central role in limiting disease severity (67–69) by rapidly responding to breakthrough infection (8) and thereby contributing to viral containment and limiting disease severity and duration. During the course of a breakthrough infection, memory T cells, formed by prior vaccination, are reactivated, rapidly expand, and respond in greater magnitude to the antigenic trigger compared to naïve T cells (70). These memory T cells usually target epitopes, which are shared with the previously encountered pathogen, while additional responses against new epitopes may also be initiated (71). In other viral infections, such as new influenza or dengue virus variants, only modest immune responses are generated against new epitopes, which may be associated with a negative clinical outcome (72). This immunological phenomenon is called “original antigenic sin” (OAS) and implies that when the epitope varies slightly, the immune system may rely on “suboptimal” memory. In the context of SARS-CoV-2, however, several studies have demonstrated that breakthrough infection leads to the induction of robust T cell responses including both pre-existing and de novo T cell responses (24, 73, 74). These responses are often directed against multiple epitopes derived from proteins that are not included in the vaccines. In addition, breakthrough infection with a viral variant elicits de novo CD8+ T cell responses not only against the infecting variant but also against newly emerging SARS-CoV-2 subvariants (74). Thus, in the case of COVID-19 vaccination, OAS does not seem to affect the robustness of the T cell response. Instead, the immunity induced by the combination of both vaccination and infection, defined as hybrid immunity, is beneficial in the case of subsequent infection. In fact, as also discussed above, the multispecific T cell response induced by hybrid immunity is especially important against emerging viral variants as they offer protection against a wider range of potential epitopes (3, 24, 73). Hybrid immunity also leads to robust and durable T cell responses in the lung mucosa (75), which have been linked to protection at the site of infection (24). Consequently, hybrid immunity induces improved protection against subsequent infection, when compared to either only infection- or vaccination-induced immunity (76, 77).
CROSS-RECOGNITION AGAINST OTHER CORONAVIRUSES OR VARIANTS OF CONCERN
Early in the pandemic, several studies reported the presence of cross-reactive T cells in individuals who had not been exposed to SARS-CoV-2. These cross-reactive memory T cells recognize epitopes of other coronaviruses, particularly of common cold coronaviruses and SARS-CoV-1 that share sequence homology with SARS-CoV-2 (12, 13, 78–80). Cross-reactivity has been described for both CD4+ and CD8+ T cells; however, it seems to be more prevalent among the CD4+ T cell compartment (81). The biological relevance of cross-reactive T cells has been shown in different scenarios. For example, their presence has been linked to a milder course of disease (33, 82) and even the prevention of SARS-CoV-2 infection in naïve individuals (83, 84). Indeed, cross-reactive T cells have been described in SARS-CoV-2 exposed healthcare workers and family members, who remained PCR- and seronegative despite viral exposure (34, 85). The potential role of cross-reactive memory CD8+ T cells as a first line of protection from infection is further supported by their presence in oropharyngeal lymphoid tissues of unexposed individuals (86). Not surprisingly, pre-existing cross-reactive T cells have also been suggested to be important after vaccination as stronger and more durable T cell responses following COVID-19 vaccination have been observed in individuals with pre-existing cross-reactive T cells (64, 87).
The most important role of cross-reactive SARS-CoV-2-specific CD8+ T cells is most likely in combating against viral variants of concerns (VOC) that emerged shortly after the initial vaccination rollout. These variants, including Alpha, Beta, Gamma, and later Omicron, exhibit several mutations that affect viral transmission, disease severity, and immune escape. The virus with the highest number of mutations, Omicron, became quickly dominant starting late 2021/early 2022 and showed enhanced evasion of humoral responses that were induced by previous infection or vaccination (88). In contrast to B cell epitopes, virus-specific T cell epitopes appear to be less affected by these mutations and are largely conserved among the different variants (4, 5) (Fig. 3). Thus, epitopes derived from VOCs can still be recognized by virus-specific CD8+ T cell responses primed by the ancestral strain (4–7, 89). In fact, although Omicron has more than 30 mutations in the spike protein (90), vaccine-induced spike-specific CD8+ T cells show high cross-reactivity (over 80%) against this variant (91, 92). This high level of cross-reactivity can be explained by several factors that make T cell epitopes less prone to mutations. One key factor is HLA polymorphism. The vast diversity of HLA genes in the human population enables the presentation and recognition of thousands of different epitopes. As a result, mutations of individual T cell epitopes would only confer an advantage to the virus in a small fraction of the population. Moreover, this diversity reduces the selective pressure on the virus to mutate these T cell epitopes. Another important factor is that the T cell epitopes are often located in regions of viral proteins that are crucial for viral function and replication (3). Hence, mutations in these regions would compromise viral fitness, making them less likely to persist (93).
Fig 3.

SARS-CoV-2-specific CD8+ T cell cross-recognition. SARS-CoV-2-specific CD8+ T cells target mainly conserved epitopes, enabling them to cross-recognize viral variants. In contrast, antibodies target surface proteins that have been more susceptible to mutation in the viral variants, leading to viral escape and reduced neutralization potency.
PATHOGENIC ROLE OF CD8+ T CELL IMMUNITY
Next to their antiviral effects, virus-specific CD8+ T cells can also contribute to disease severity, e.g., a more severe clinical course of COVID-19 (94–96). The exact mechanisms contributing to a more pronounced pathogenic effect of SARS-CoV-2-specific CD8+ T cells are not well understood. It is possible that they may indirectly contribute to the pathogenesis of the disease by, e.g., delayed induction and reduced frequencies (19, 39–41, 97), most likely leading to failure to appropriately limit viral replication (Fig. 4). In addition, impaired viral control in the early stages of infection causes increased viral replication that results in an enhanced and dysregulated immune response later in the infection. In line with this, delayed, but prolonged and excessive activation of CD8+ T cells has been observed in patients with severe COVID-19 (94, 95, 97). Furthermore, CD8+ T cells can also directly contribute to pathology by exerting their effector functions in an uncontrolled and nonspecific manner (immunopathology; Fig. 4). During viral infection, CD8+ T cells rapidly produce proinflammatory cytokines upon antigen encounter to mediate efficient viral clearance. However, if these effector functions are unrestricted, they can cause tissue damage and inflammation. Indeed, a higher frequency of bronchoalveolar lavage cytotoxic CD8+ T cells has been linked to epithelial damage and airway disease in individuals with persistent symptoms after acute COVID-19 (98). Moreover, tissue damage can also be caused by misdirected cytotoxicity of non-SARS-CoV-2-specific CD8+ T cell activation that provokes apoptosis of uninfected cells and, thus, may also contribute to the pathogenesis of severe disease (Fig. 4). This TCR-independent T cell activation can be induced, e.g., by the complement system (99) or by a proinflammatory environment (100). In the latter case, the non-SARS-CoV-2-specific T cells are readily activated in a TCR-independent, bystander manner by proinflammatory cytokines, such as type I IFNs (100). Due to their rapid induction, bystander-activated T cells can be present even before the induction and development of antigen-specific T cell responses, which usually takes a few days. In this way, bystander-activated T cells may play an important role in the early viral defense. Yet, in cases of excessive and uncontrolled stimulation, they may also contribute to immunopathology. These bystander-activated T cells can be identified by the expression of activation markers (e.g., CD38 and HLA-DR), cytotoxic molecules (e.g., granzyme B), and the natural killer cell-activating receptor NKG2D (101). These cells have been found in patients with severe COVID-19 (97), supporting their possible role in mediating disease severity. Furthermore, CD8+ T cells have also been suggested to be involved in the pathogenesis of long COVID. However, findings on this topic are conflicting. Some studies have reported the presence of exhausted SARS-CoV-2-specific CD8+ T cells in individuals who suffer from long COVID (102), while others have identified hyperactivated CD8+ T cells with augmented granzyme B and IFN-y production (103, 104). In contrast to these findings, another in-depth study observed similar SARS-CoV-2-specific T cell responses, memory formation, and TCRαβ clonotypes in people with long COVID compared to non-long COVID individuals (105), indicating that SARS-CoV-2-specific T cells may not play a significant role in the pathogenesis of long COVID. Alternatively, it has been hypothesized that immune responses to recently reactivated, latently persisting viruses, such as EBV, may be involved in the development of long COVID symptoms (106). A further possible hypothesis is that immunosuppressive KIR+ CD8+ T cells may contribute to disease pathogenesis by hampering virus-specific immune responses. Indeed, higher frequencies of KIR+ CD8+ T cells have been detected in patients with severe COVID-19 (107). Nevertheless, despite extensive research, the exact role of CD8+ T cells in long COVID remains to be determined.
Fig 4.

Protective and pathological role of CD8+ T cells in acute viral infection. Early and high frequencies of SARS-CoV-2-specific CD8+ T cell responses mediate rapid viral clearance, resulting in protection from severe disease (left). Delayed and reduced frequencies of SARS-CoV-2-specific CD8+ T cell responses fail to appropriately control viral replication and spread, leading to virus-induced pathology (middle). Nonspecific and uncontrolled effector functions lead to the killing of uninfected cells, but not to viral control, resulting in tissue damage (right).
CONCLUSIONS AND OPEN QUESTIONS
SARS-CoV-2 has been an excellent model for studying antiviral CD8+ T cell immunity in humans, confirming fundamental principles of CD8+ T cell responses in humans, while also revealing novel aspects of their role in acute viral infection. Even though much has been learned, several key questions still remain unanswered. For instance, memory CD8+ T cells with stem cell-like characteristics have been detected in the peripheral blood after natural infection and vaccination. So far, these memory CD8+ T cells have only been studied for 12 months after infection and 6–8 months after vaccination. Nevertheless, it can be assumed that SARS-CoV-2 induces a similarly long-lasting T cell immunity, as described after SARS-CoV-1 infection, where virus-specific T cells were still detectable 17 years after infection (80). However, as SARS-CoV-2 persists as an endemic pathogen, the question remains open as to how this CD8+ T cell immunity is shaped and maintained in terms of repertoire and function by frequent exposure to the virus. Furthermore, the factors that influence the kinetics and magnitude of SARS-CoV-2-specific T cell responses and their dysregulation during severe disease still need to be determined. The complex interaction of CD8+ T cells with other components of the immune system, e.g., with the innate immune system, is also only little understood. Finally, the role of CD8+ T cells in the pathogenesis of long COVID remains a crucial area of investigation, and future studies need to reveal whether long COVID is caused by prolonged activation of immune cells due to dysregulation, antigen persistence, or by autoimmune phenomena. In conclusion, SARS-CoV-2 continues to serve as a valuable model for studying CD8+ T cell immunity in humans and remains especially pertinent for the development of vaccine strategies for other emerging viruses.
ACKNOWLEDGMENTS
This work was funded by grants from the German Research Foundation (CRC1160 - Project ID: 256073931—A02 to R.T. and M.H.; TRR179 - Project ID: 272983813—TP01 to R.T. and TP20 to M.H.; and CRC1479 - Project ID: 441891347—P10 to M.H.). This work was also supported by the project “Virological and immunological determinants of COVID-19 pathogenesis – lessons to get prepared for future pandemics (KA1-Co-02 “COVIPA”)”, a grant from the Helmholtz Association’s Initiative and Networking Fund to R.T. and M.H. M.H. was further supported by the DFG Heisenberg program of the German Research Foundation (project no. HO 5836/2-1).
Illustrations were created with BioRender.com. We thank Julia Lang-Meli and David Reeg for helpful input. Finally, we apologize for the omission of papers that are not discussed here due to space limitations.
Contributor Information
Robert Thimme, Email: robert.thimme@uniklinik-freiburg.de.
David E. Levy, New York University Department of Microbiology, New York, New York, USA
REFERENCES
- 1. Bange EM, Han NA, Wileyto P, Kim JY, Gouma S, Robinson J, Greenplate AR, Hwee MA, Porterfield F, Owoyemi O, et al. 2021. CD8+ T cells contribute to survival in patients with COVID-19 and hematologic cancer. Nat Med 27:1280–1289. doi: 10.1038/s41591-021-01386-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Liu J, Yu J, McMahan K, Jacob-Dolan C, He X, Giffin V, Wu C, Sciacca M, Powers O, Nampanya F, Miller J, Lifton M, Hope D, Hall K, Hachmann NP, Chung B, Anioke T, Li W, Muench J, Gamblin A, Boursiquot M, Cook A, Lewis MG, Andersen H, Barouch DH. 2022. CD8 T cells contribute to vaccine protection against SARS-CoV-2 in macaques. Sci Immunol 7:eabq7647. doi: 10.1126/sciimmunol.abq7647 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Lang-Meli J, Luxenburger H, Wild K, Karl V, Oberhardt V, Salimi Alizei E, Graeser A, Reinscheid M, Roehlen N, Reeg DB, et al. 2022. SARS-CoV-2-specific T-cell epitope repertoire in convalescent and mRNA-vaccinated individuals. Nat Microbiol 7:675–679. doi: 10.1038/s41564-022-01106-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Tarke A, Sidney J, Methot N, Yu ED, Zhang Y, Dan JM, Goodwin B, Rubiro P, Sutherland A, Wang E, Frazier A, Ramirez SI, Rawlings SA, Smith DM, da Silva Antunes R, Peters B, Scheuermann RH, Weiskopf D, Crotty S, Grifoni A, Sette A. 2021. 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: 10.1016/j.xcrm.2021.100355 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Redd AD, Nardin A, Kared H, Bloch EM, Abel B, Pekosz A, Laeyendecker O, Fehlings M, Quinn TC, Tobian AAR. 2022. Minimal crossover between mutations associated with omicron variant of SARS-CoV-2 and CD8+ T-Cell epitopes identified in COVID-19 convalescent individuals. mBio:e03617-21. doi: 10.1128/mbio.03617-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Redd AD, Nardin A, Kared H, Bloch EM, Pekosz A, Laeyendecker O, Abel B, Fehlings M, Quinn TC, 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:fab143. doi: 10.1093/ofid/ofab143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Tarke A, Coelho CH, Zhang Z, Dan JM, Yu ED, Methot N, Bloom NI, Goodwin B, Phillips E, Mallal S, Sidney J, Filaci G, Weiskopf D, da Silva Antunes R, Crotty S, Grifoni A, Sette A. 2022. SARS-CoV-2 vaccination induces immunological T cell memory able to cross-recognize variants from Alpha to Omicron. Cell 185:847–859. doi: 10.1016/j.cell.2022.01.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Reinscheid M, Luxenburger H, Karl V, Graeser A, Giese S, Ciminski K, Reeg DB, Oberhardt V, Roehlen N, Lang-Meli J, Heim K, Gross N, Baum C, Rieg S, Speer C, Emmerich F, Breisinger S, Steinmann D, Bengsch B, Boettler T, Kochs G, Schwemmle M, Thimme R, Neumann-Haefelin C, Hofmann M. 2022. COVID-19 mRNA booster vaccine induces transient CD8+ T effector cell responses while conserving the memory pool for subsequent reactivation. Nat Commun 13:4631. doi: 10.1038/s41467-022-32324-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Oberhardt V, Luxenburger H, Kemming J, Schulien I, Ciminski K, Giese S, Csernalabics B, Lang-Meli J, Janowska I, Staniek J, et al. 2021. Rapid and stable mobilization of CD8+ T cells by SARS-CoV-2 mRNA vaccine. Nature 597:268–273. doi: 10.1038/s41586-021-03841-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Cohen KW, Linderman SL, Moodie Z, Czartoski J, Lai L, Mantus G, Norwood C, Nyhoff LE, Edara VV, Floyd K, et al. 2021. Longitudinal analysis shows durable and broad immune memory after SARS-CoV-2 infection with persisting antibody responses and memory B and T cells. Cell Rep Med 2:100354. doi: 10.1016/j.xcrm.2021.100354 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Dan JM, Mateus J, Kato Y, Hastie KM, Yu ED, Faliti CE, Grifoni A, Ramirez SI, Haupt S, Frazier A, Nakao C, Rayaprolu V, Rawlings SA, Peters B, Krammer F, Simon V, Saphire EO, Smith DM, Weiskopf D, Sette A, Crotty S. 2021. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science 371:eabf4063. doi: 10.1126/science.abf4063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Schulien I, Kemming J, Oberhardt V, Wild K, Seidel LM, Killmer S, 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: 10.1038/s41591-020-01143-2 [DOI] [PubMed] [Google Scholar]
- 13. 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, 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. doi: 10.1016/j.cell.2020.05.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Rha MS, Jeong HW, Ko JH, Choi SJ, Seo IH, Lee JS, Sa M, Kim AR, Joo EJ, Ahn JY, Kim JH, Song KH, Kim ES, Oh DH, Ahn MY, Choi HK, Jeon JH, Choi JP, Kim HB, Kim YK, Park SH, Choi WS, Choi JY, Peck KR, Shin EC. 2021. PD-1-expressing SARS-CoV-2-specific CD8+ T cells are not exhausted, but functional in patients with COVID-19. Immunity 54:44–52. doi: 10.1016/j.immuni.2020.12.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Tarke A, Sidney J, Kidd CK, Dan JM, Ramirez SI, Yu ED, Mateus J, da Silva Antunes R, Moore E, Rubiro P, Methot N, Phillips E, Mallal S, Frazier A, Rawlings SA, Greenbaum JA, Peters B, Smith DM, Crotty S, Weiskopf D, Grifoni A, Sette A. 2021. Comprehensive analysis of T cell immunodominance and immunoprevalence of SARS-CoV-2 epitopes in COVID-19 cases. Cell Rep Med 2:100204. doi: 10.1016/j.xcrm.2021.100204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Cheon IS, Li C, Son YM, Goplen NP, Wu Y, Cassmann T, Wang Z, Wei X, Tang J, Li Y, et al. 2021. Immune signatures underlying post-acute COVID-19 lung sequelae. Sci Immunol 6:eabk1741. doi: 10.1126/sciimmunol.abk1741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Grau-Expósito J, Sánchez-Gaona N, Massana N, Suppi M, Astorga-Gamaza A, Perea D, Rosado J, Falcó A, Kirkegaard C, Torrella A, et al. 2021. Peripheral and lung resident memory T cell responses against SARS-CoV-2. Nat Commun 12:3010. doi: 10.1038/s41467-021-23333-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Szabo PA, Dogra P, Gray JI, Wells SB, Connors TJ, Weisberg SP, Krupska I, Matsumoto R, Poon MML, Idzikowski E, Morris SE, Pasin C, Yates AJ, Ku A, Chait M, Davis-Porada J, Guo XV, Zhou J, Steinle M, Mackay S, Saqi A, Baldwin MR, Sims PA, Farber DL. 2021. Longitudinal profiling of respiratory and systemic immune responses reveals myeloid cell-driven lung inflammation in severe COVID-19. Immunity 54:797–814. doi: 10.1016/j.immuni.2021.03.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Liao M, Liu Y, Yuan J, Wen Y, Xu G, Zhao J, Cheng L, Li J, Wang X, Wang F, Liu L, Amit I, Zhang S, Zhang Z. 2020. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat Med 26:842–844. doi: 10.1038/s41591-020-0901-9 [DOI] [PubMed] [Google Scholar]
- 20. Poon MML, Rybkina K, Kato Y, Kubota M, Matsumoto R, Bloom NI, Zhang Z, Hastie KM, Grifoni A, Weiskopf D, Wells SB, Ural BB, Lam N, Szabo PA, Dogra P, Lee YS, Gray JI, Bradley MC, Brusko MA, Brusko TM, Saphire EO, Connors TJ, Sette A, Crotty S, Farber DL. 2021. SARS-CoV-2 infection generates tissue-localized immunological memory in humans. Sci Immunol 6:eabl9105. doi: 10.1126/sciimmunol.abl9105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Roukens AHE, Pothast CR, König M, Huisman W, Dalebout T, Tak T, Azimi S, Kruize Y, Hagedoorn RS, Zlei M, et al. 2022. Prolonged activation of nasal immune cell populations and development of tissue-resident SARS-CoV-2-specific CD8+ T cell responses following COVID-19. Nat Immunol 23:23–32. doi: 10.1038/s41590-021-01095-w [DOI] [PubMed] [Google Scholar]
- 22. Rha MS, Kim G, Lee S, Kim J, Jeong Y, Jung CM, Noh HE, Noh JY, Kim YM, Cho HJ, Kim CH, Shin EC. 2024. SARS-CoV-2 spike-specific nasal-resident CD49a+CD8+ memory T cells exert immediate effector functions with enhanced IFN-γ production. Nat Commun 15:8355. doi: 10.1038/s41467-024-52689-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Ramirez SI, Faraji F, Hills LB, Lopez PG, Goodwin B, Stacey HD, Sutton HJ, Hastie KM, Saphire EO, Kim HJ, Mashoof S, Yan CH, DeConde AS, Levi G, Crotty S. 2024. Immunological memory diversity in the human upper airway. Nature 632:630–636. doi: 10.1038/s41586-024-07748-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Lim JME, Tan AT, Le Bert N, Hang SK, Low JGH, Bertoletti A. 2022. SARS-CoV-2 breakthrough infection in vaccinees induces virus-specific nasal-resident CD8+ and CD4+ T cells of broad specificity. J Exp Med 219:e20220780. doi: 10.1084/jem.20220780 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Neidleman J, Luo X, Frouard J, Xie G, Gill G, Stein ES, McGregor M, Ma T, George AF, Kosters A, Greene WC, Vasquez J, Ghosn E, Lee S, Roan NR. 2020. SARS-CoV-2-specific T cells exhibit phenotypic features of helper function, lack of terminal differentiation, and high proliferation potential. Cell Rep Med 1:100081. doi: 10.1016/j.xcrm.2020.100081 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Sridhar S, Begom S, Bermingham A, Hoschler K, Adamson W, Carman W, Bean T, Barclay W, Deeks JJ, Lalvani A. 2013. Cellular immune correlates of protection against symptomatic pandemic influenza. Nat Med 19:1305–1312. doi: 10.1038/nm.3350 [DOI] [PubMed] [Google Scholar]
- 27. Lilleri D, Fornara C, Revello MG, Gerna G. 2008. Human cytomegalovirus-specific memory CD8+ and CD4+ T cell differentiation after primary infection. J Infect Dis 198:536–543. doi: 10.1086/590118 [DOI] [PubMed] [Google Scholar]
- 28. Dunne PJ, Faint JM, Gudgeon NH, Fletcher JM, Plunkett FJ, Soares MVD, Hislop AD, Annels NE, Rickinson AB, Salmon M, Akbar AN. 2002. Epstein-Barr virus-specific CD8(+) T cells that re-express CD45RA are apoptosis-resistant memory cells that retain replicative potential. Blood 100:933–940. doi: 10.1182/blood-2002-01-0160 [DOI] [PubMed] [Google Scholar]
- 29. Northfield JW, Loo CP, Barbour JD, Spotts G, Hecht FM, Klenerman P, Nixon DF, Michaëlsson J. 2007. Human immunodeficiency virus type 1 (HIV-1)-specific CD8+ T(EMRA) cells in early infection are linked to control of HIV-1 viremia and predict the subsequent viral load set point. J Virol 81:5759–5765. doi: 10.1128/JVI.00045-07 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Lu Z, Laing ED, Pena DaMata J, Pohida K, Tso MS, Samuels EC, Epsi NJ, Dorjbal B, Lake C, Richard SA, et al. 2021. Durability of SARS-CoV-2-Specific T-cell responses at 12 months postinfection. J Infect Dis:2010–2019. doi: 10.1101/2021.08.11.455984 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Adamo S, Michler J, Zurbuchen Y, Cervia C, Taeschler P, Raeber ME, Baghai Sain S, Nilsson J, Moor AE, Boyman O. 2022. Signature of long-lived memory CD8+ T cells in acute SARS-CoV-2 infection. Nature 602:148–155. doi: 10.1038/s41586-021-04280-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Rydyznski Moderbacher C, Ramirez SI, Dan JM, Grifoni A, Hastie KM, Weiskopf D, Belanger S, Abbott RK, Kim C, Choi J, Kato Y, Crotty EG, Kim C, Rawlings SA, Mateus J, Tse LPV, Frazier A, Baric R, Peters B, Greenbaum J, Ollmann Saphire E, Smith DM, Sette A, Crotty S. 2020. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell 183:996–1012. doi: 10.1016/j.cell.2020.09.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Peng Y, Felce SL, Dong D, Penkava F, Mentzer AJ, Yao X, Liu G, Yin Z, Chen J-L, Lu Y, et al. 2022. 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: 10.1038/s41590-021-01084-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. 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. doi: 10.1016/j.cell.2020.08.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Ramirez SI, Lopez PG, Faraji F, Parikh UM, Heaps A, Ritz J, Moser C, Eron JJ, Wohl D, Currier J, Daar ES, Greninger A, Klekotka P, Grifoni A, Weiskopf D, Sette A, Peters B, Hughes MD, Chew KW, Smith DM, Crotty S, Accelerating C-T, Vaccines AST. 2024. Early antiviral CD4+ and CD8+ T cells are associated with upper airway clearance of SARS-CoV-2. JCI Insight 9:e186078. doi: 10.1172/jci.insight.186078 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Le Bert N, Clapham HE, Tan AT, Chia WN, Tham CYL, Lim JM, Kunasegaran K, Tan LWL, Dutertre CA, Shankar N, et al. 2021. Highly functional virus-specific cellular immune response in asymptomatic SARS-CoV-2 infection. J Exp Med 218:e20202617. doi: 10.1084/jem.20202617 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Lindeboom RGH, Worlock KB, Dratva LM, Yoshida M, Scobie D, Wagstaffe HR, Richardson L, Wilbrey-Clark A, Barnes JL, Kretschmer L, et al. 2024. Human SARS-CoV-2 challenge uncovers local and systemic response dynamics. Nature 631:189–198. doi: 10.1038/s41586-024-07575-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Wang Z, Yang X, Zhong J, Zhou Y, Tang Z, Zhou H, He J, Mei X, Tang Y, Lin B, Chen Z, McCluskey J, Yang J, Corbett AJ, Ran P. 2021. Exposure to SARS-CoV-2 generates T-cell memory in the absence of a detectable viral infection. Nat Commun 12:1724. doi: 10.1038/s41467-021-22036-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Mathew D, Giles JR, Baxter AE, Oldridge DA, Greenplate AR, Wu JE, Alanio C, Kuri-Cervantes L, Pampena MB, D’Andrea K, et al. 2020. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 369. doi: 10.1126/science.abc8511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Tan AT, Linster M, Tan CW, Le Bert N, Chia WN, Kunasegaran K, Zhuang Y, Tham CYL, Chia A, Smith GJD, Young B, Kalimuddin S, Low JGH, Lye D, Wang LF, Bertoletti A. 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: 10.1016/j.celrep.2021.108728 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Melms JC, Biermann J, Huang H, Wang Y, Nair A, Tagore S, Katsyv I, Rendeiro AF, Amin AD, Schapiro D, et al. 2021. A molecular single-cell lung atlas of lethal COVID-19. Nature 595:114–119. doi: 10.1038/s41586-021-03569-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. McMahan K, Yu J, Mercado NB, Loos C, Tostanoski LH, Chandrashekar A, Liu J, Peter L, Atyeo C, Zhu A, et al. 2021. Correlates of protection against SARS-CoV-2 in rhesus macaques. Nature 590:630–634. doi: 10.1038/s41586-020-03041-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Lim JME, Hang SK, Hariharaputran S, Chia A, Tan N, Lee ES, Chng E, Lim PL, Young BE, Lye DC, Le Bert N, Bertoletti A, Tan AT. 2022. A comparative characterization of SARS-CoV-2-specific T cells induced by mRNA or inactive virus COVID-19 vaccines. Cell Rep Med 3:100793. doi: 10.1016/j.xcrm.2022.100793 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Bueno SM, Abarca K, González PA, Gálvez NMS, Soto JA, Duarte LF, Schultz BM, Pacheco GA, González LA, Vázquez Y, et al. 2022. Safety and immunogenicity of an inactivated severe acute respiratory syndrome Coronavirus 2 vaccine in a subgroup of healthy adults in Chile. Clin Infect Dis 75:e792–e804. doi: 10.1093/cid/ciab823 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Vikkurthi R, Ansari A, Pai AR, Jha SN, Sachan S, Pandit S, Nikam B, Kalia A, Jit BP, Parray HA, et al. 2022. Inactivated whole-virion vaccine BBV152/Covaxin elicits robust cellular immune memory to SARS-CoV-2 and variants of concern. Nat Microbiol 7:974–985. doi: 10.1038/s41564-022-01161-5 [DOI] [PubMed] [Google Scholar]
- 46. Barbeau DJ, Martin JM, Carney E, Dougherty E, Doyle JD, Dermody TS, Hoberman A, Williams JV, Michaels MG, Alcorn JF, Paul Duprex W, McElroy AK. 2022. Comparative analysis of human immune responses following SARS-CoV-2 vaccination with BNT162b2, mRNA-1273, or Ad26.COV2.S. NPJ Vaccines 7:77. doi: 10.1038/s41541-022-00504-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Maringer Y, Nelde A, Schroeder SM, Schuhmacher J, Hörber S, Peter A, Karbach J, Jäger E, Walz JS. 2022. Durable spike-specific T cell responses after different COVID-19 vaccination regimens are not further enhanced by booster vaccination. Sci Immunol 7:eadd3899. doi: 10.1126/sciimmunol.add3899 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Sahin U, Muik A, Derhovanessian E, Vogler I, Kranz LM, Vormehr M, Baum A, Pascal K, Quandt J, Maurus D, et al. 2020. COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T cell responses. Nature New Biol 586:594–599. doi: 10.1038/s41586-020-2814-7 [DOI] [PubMed] [Google Scholar]
- 49. 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: 10.1038/s41591-021-01507-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Sabatino JJ Jr, Mittl K, Rowles WM, McPolin K, Rajan JV, Laurie MT, Zamecnik CR, Dandekar R, Alvarenga BD, Loudermilk RP, Gerungan C, Spencer CM, Sagan SA, Augusto DG, Alexander JR, DeRisi JL, Hollenbach JA, Wilson MR, Zamvil SS, Bove R. 2022. Multiple sclerosis therapies differentially affect SARS-CoV-2 vaccine–induced antibody and T cell immunity and function. JCI Insight 7. doi: 10.1172/jci.insight.156978 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Luxenburger H, Reeg DB, Lang-Meli J, Reinscheid M, Eisner M, Bettinger D, Oberhardt V, Salimi Alizei E, Wild K, Graeser A, Karl V, Emmerich F, Klein F, Panning M, Huzly D, Bengsch B, Boettler T, Elling R, Thimme R, Hofmann M, Neumann-Haefelin C. 2023. Boosting compromised SARS-CoV-2-specific immunity with mRNA vaccination in liver transplant recipients. J Hepatol 78:1017–1027. doi: 10.1016/j.jhep.2023.02.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Riise J, Meyer S, Blaas I, Chopra A, Tran TT, Delic-Sarac M, Hestdalen ML, Brodin E, Rustad EH, Dai K-Z, Vaage JT, Nissen-Meyer LSH, Sund F, Wader KF, Bjornevik AT, Meyer PA, Nygaard GO, König M, Smeland S, Lund-Johansen F, Olweus J, Kolstad A. 2022. Rituximab-treated patients with lymphoma develop strong CD8 T-cell responses following COVID-19 vaccination. Br J Haematol 197:697–708. doi: 10.1111/bjh.18149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Gao Y, Cai C, Wullimann D, Niessl J, Rivera-Ballesteros O, Chen P, Lange J, Cuapio A, Blennow O, Hansson L, et al. 2022. Immunodeficiency syndromes differentially impact the functional profile of SARS-CoV-2-specific T cells elicited by mRNA vaccination. Immunity 55:1732–1746. doi: 10.1016/j.immuni.2022.07.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Kalimuddin S, Tham CYL, Qui M, de Alwis R, Sim JXY, Lim JME, Tan H-C, Syenina A, Zhang SL, Le Bert N, Tan AT, Leong YS, Yee JX, Ong EZ, Ooi EE, Bertoletti A, Low JG. 2021. Early T cell and binding antibody responses are associated with COVID-19 RNA vaccine efficacy onset. Med 2:682–688. doi: 10.1016/j.medj.2021.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Ishii H, Nomura T, Yamamoto H, Nishizawa M, Thu Hau TT, Harada S, Seki S, Nakamura-Hoshi M, Okazaki M, Daigen S, Kawana-Tachikawa A, Nagata N, Iwata-Yoshikawa N, Shiwa N, Suzuki T, Park ES, Ken M, Onodera T, Takahashi Y, Kusano K, Shimazaki R, Suzaki Y, Ami Y, Matano T. 2022. Neutralizing-antibody-independent SARS-CoV-2 control correlated with intranasal-vaccine-induced CD8+ T cell responses. Cell Rep Med 3:100520. doi: 10.1016/j.xcrm.2022.100520 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Chandrashekar A, Yu J, McMahan K, Jacob-Dolan C, Liu J, He X, Hope D, Anioke T, Barrett J, Chung B, et al. 2022. Vaccine protection against the SARS-CoV-2 Omicron variant in macaques. Cell 185:1549–1555. doi: 10.1016/j.cell.2022.03.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Atmar RL, Lyke KE, Deming ME, Jackson LA, Branche AR, El Sahly HM, Rostad CA, Martin JM, Johnston C, Rupp RE, et al. 2022. Homologous and heterologous Covid-19 booster vaccinations. N Engl J Med 386:1046–1057. doi: 10.1056/NEJMoa2116414 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Schmidt T, Klemis V, Schub D, Mihm J, Hielscher F, Marx S, Abu-Omar A, Ziegler L, Guckelmus C, Urschel R, Schneitler S, Becker SL, Gärtner BC, Sester U, Sester M. 2021. Immunogenicity and reactogenicity of heterologous ChAdOx1 nCoV-19/mRNA vaccination. Nat Med 27:1530–1535. doi: 10.1038/s41591-021-01464-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Asante MA, Michelsen ME, Balakumar MM, Kumburegama B, Sharifan A, Thomsen AR, Korang SK, Gluud C, Menon S. 2024. Heterologous versus homologous COVID-19 booster vaccinations for adults: systematic review with meta-analysis and trial sequential analysis of randomised clinical trials. BMC Med 22:263. doi: 10.1186/s12916-024-03471-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Akondy RS, Fitch M, Edupuganti S, Yang S, Kissick HT, Li KW, Youngblood BA, Abdelsamed HA, McGuire DJ, Cohen KW, Alexe G, Nagar S, McCausland MM, Gupta S, Tata P, Haining WN, McElrath MJ, Zhang D, Hu B, Greenleaf WJ, Goronzy JJ, Mulligan MJ, Hellerstein M, Ahmed R. 2017. Origin and differentiation of human memory CD8 T cells after vaccination. Nature 552:362–367. doi: 10.1038/nature24633 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Guerrera G, Picozza M, D’Orso S, Placido R, Pirronello M, Verdiani A, Termine A, Fabrizio C, Giannessi F, Sambucci M, Balice MP, Caltagirone C, Salvia A, Rossini A, Battistini L, Borsellino G. 2021. BNT162b2 vaccination induces durable SARS-CoV-2-specific T cells with a stem cell memory phenotype. Sci Immunol 6:eabl5344. doi: 10.1126/sciimmunol.abl5344 [DOI] [PubMed] [Google Scholar]
- 62. Barouch DH, Stephenson KE, Sadoff J, Yu J, Chang A, Gebre M, McMahan K, Liu J, Chandrashekar A, Patel S, Le Gars M, de Groot AM, Heerwegh D, Struyf F, Douoguih M, van Hoof J, Schuitemaker H. 2021. Durable humoral and cellular immune responses 8 months after Ad26.COV2.S vaccination. N Engl J Med 385:951–953. doi: 10.1056/NEJMc2108829 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Zhang Z, Mateus J, Coelho CH, Dan JM, Moderbacher CR, Gálvez RI, Cortes FH, Grifoni A, Tarke A, Chang J, Escarrega EA, Kim C, Goodwin B, Bloom NI, Frazier A, Weiskopf D, Sette A, Crotty S. 2022. Humoral and cellular immune memory to four COVID-19 vaccines. Cell 185:2434–2451. doi: 10.1016/j.cell.2022.05.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Mateus J, Dan JM, Zhang Z, Rydyznski Moderbacher C, Lammers M, Goodwin B, Sette A, Crotty S, Weiskopf D. 2021. Low-dose mRNA-1273 COVID-19 vaccine generates durable memory enhanced by cross-reactive T cells. Science 374:eabj9853. doi: 10.1126/science.abj9853 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Tober-Lau P, Schwarz T, Vanshylla K, Hillus D, Gruell H, Suttorp N, Landgraf I, Kappert K, Seybold J, Drosten C, Klein F, Kurth F, Sander LE, Corman VM. 2021. Long-term immunogenicity of BNT162b2 vaccination in older people and younger health-care workers. Lancet Respir Med 9:e104–e105. doi: 10.1016/S2213-2600(21)00456-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Demaret J, Corroyer-Simovic B, Alidjinou EK, Goffard A, Trauet J, Miczek S, Vuotto F, Dendooven A, Huvent-Grelle D, Podvin J, Dreuil D, Faure K, Deplanque D, Bocket L, Duhamel A, Labreuche J, Sobaszek A, Hisbergues M, Puisieux F, Labalette M, Lefèvre G. 2021. Impaired functional T-cell response to SARS-CoV-2 after two doses of BNT162b2 mRNA vaccine in older people. Front Immunol 12:778679. doi: 10.3389/fimmu.2021.778679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Rovida F, Cassaniti I, Paolucci S, Percivalle E, Sarasini A, Piralla A, Giardina F, Sammartino JC, Ferrari A, Bergami F, Muzzi A, Novelli V, Meloni A, Cutti S, Grugnetti AM, Grugnetti G, Rona C, Daglio M, Marena C, Triarico A, Lilleri D, Baldanti F. 2021. SARS-CoV-2 vaccine breakthrough infections with the alpha variant are asymptomatic or mildly symptomatic among health care workers. Nat Commun 12:6032. doi: 10.1038/s41467-021-26154-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Rümke LW, Groenveld FC, van Os YMG, Praest P, Tanja AAN, de Jong DTCM, Symons J, Schuurman R, Reinders T, Hofstra LM, Nierkens S, Thijsen SFT, Heron M, Lebbink R-J, Beekman JM, Nijhuis M, Wensing AMJ. 2022. In-depth Characterization of vaccine breakthrough infections with SARS-CoV-2 among health care workers in a Dutch academic medical center. Open Forum Infect Dis 9:ofab553. doi: 10.1093/ofid/ofab553 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Paniskaki K, Anft M, Meister TL, Marheinecke C, Pfaender S, Skrzypczyk S, Seibert FS, Thieme CJ, Konik MJ, Dolff S, Anastasiou O, Holzer B, Dittmer U, Queren C, Fricke L, Rohn H, Westhoff TH, Witzke O, Stervbo U, Roch T, Babel N. 2022. Immune response in moderate to critical breakthrough COVID-19 infection after mRNA vaccination. Front Immunol 13:816220. doi: 10.3389/fimmu.2022.816220 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Painter MM, Johnston TS, Lundgreen KA, Santos JJS, Qin JS, Goel RR, Apostolidis SA, Mathew D, Fulmer B, Williams JC, et al. 2023. Prior vaccination promotes early activation of memory T cells and enhances immune responses during SARS-CoV-2 breakthrough infection. Nat Immunol 24:1711–1724. doi: 10.1038/s41590-023-01613-y [DOI] [PubMed] [Google Scholar]
- 71. Tarke A, Ramezani-Rad P, Alves Pereira Neto T, Lee Y, Silva-Moraes V, Goodwin B, Bloom N, Siddiqui L, Avalos L, Frazier A, Zhang Z, da Silva Antunes R, Dan J, Crotty S, Grifoni A, Sette A. 2024. SARS-CoV-2 breakthrough infections enhance T cell response magnitude, breadth, and epitope repertoire. Cell Rep Med 5:101583. doi: 10.1016/j.xcrm.2024.101583 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Zhang A, Stacey HD, Mullarkey CE, Miller MS. 2019. Original antigenic sin: how first exposure shapes lifelong anti-influenza virus immune responses. J Immunol 202:335–340. doi: 10.4049/jimmunol.1801149 [DOI] [PubMed] [Google Scholar]
- 73. Minervina AA, Pogorelyy MV, Kirk AM, Crawford JC, Allen EK, Chou C-H, Mettelman RC, Allison KJ, Lin C-Y, Brice DC, et al. 2022. SARS-CoV-2 antigen exposure history shapes phenotypes and specificity of memory CD8+ T cells. Nat Immunol 23:781–790. doi: 10.1038/s41590-022-01184-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Kim SH, Kim J, Jung S, Noh JY, Kim J, Park H, Song YG, Peck KR, Park SH, Park MS, Ko JH, Song JY, Choi JY, Jung MK, Shin EC. 2024. Omicron BA.2 breakthrough infection elicits CD8+ T cell responses recognizing the spike of later Omicron subvariants. Sci Immunol 9:eade6132. doi: 10.1126/sciimmunol.ade6132 [DOI] [PubMed] [Google Scholar]
- 75. Mitsi E, Diniz MO, Reiné J, Collins AM, Robinson RE, Hyder-Wright A, Farrar M, Liatsikos K, Hamilton J, Onyema O, Urban BC, Solórzano C, Belij-Rammerstorfer S, Sheehan E, Lambe T, Draper SJ, Weiskopf D, Sette A, Maini MK, Ferreira DM. 2023. Respiratory mucosal immune memory to SARS-CoV-2 after infection and vaccination. Nat Commun 14:6815. doi: 10.1038/s41467-023-42433-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Ntziora F, Kostaki EG, Karapanou A, Mylona M, Tseti I, Sipsas NV, Paraskevis D, Sfikakis PP. 2022. Protection of vaccination versus hybrid immunity against infection with COVID-19 Omicron variants among Health-Care Workers. Vaccine (Auckl) 40:7195–7200. doi: 10.1016/j.vaccine.2022.09.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Altarawneh HN, Chemaitelly H, Ayoub HH, Tang P, Hasan MR, Yassine HM, Al-Khatib HA, Smatti MK, Coyle P, Al-Kanaani Z, Al-Kuwari E, Jeremijenko A, Kaleeckal AH, Latif AN, Shaik RM, Abdul-Rahim HF, Nasrallah GK, Al-Kuwari MG, Butt AA, Al-Romaihi HE, Al-Thani MH, Al-Khal A, Bertollini R, Abu-Raddad LJ. 2022. Effects of previous infection and vaccination on symptomatic Omicron infections. N Engl J Med 387:21–34. doi: 10.1056/NEJMoa2203965 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. 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: 10.1038/s41590-020-00808-x [DOI] [PubMed] [Google Scholar]
- 79. Prakash S, Srivastava R, Coulon PG, Dhanushkodi NR, Chentoufi AA, Tifrea DF, Edwards RA, Figueroa CJ, Schubl SD, Hsieh L, Buchmeier MJ, Bouziane M, Nesburn AB, Kuppermann BD, BenMohamed L. 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: 10.4049/jimmunol.2001438 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Le Bert N, Tan AT, Kunasegaran K, Tham CYL, Hafezi M, Chia A, Chng MHY, Lin M, Tan N, Linster M, Chia WN, Chen MI-C, Wang L-F, Ooi EE, Kalimuddin S, Tambyah PA, Low JG-H, Tan Y-J, Bertoletti A. 2020. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 584:457–462. doi: 10.1038/s41586-020-2550-z [DOI] [PubMed] [Google Scholar]
- 81. Ferretti AP, Kula T, Wang Y, Nguyen DMV, Weinheimer A, Dunlap GS, Xu Q, Nabilsi N, Perullo CR, Cristofaro AW, Whitton HJ, Virbasius A, Olivier KJ Jr, Buckner LR, Alistar AT, Whitman ED, Bertino SA, Chattopadhyay S, MacBeath G. 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. doi: 10.1016/j.immuni.2020.10.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Sagar M, Reifler K, Rossi M, Miller NS, Sinha P, White LF, Mizgerd JP. 2021. Recent endemic coronavirus infection is associated with less-severe COVID-19. J Clin Invest 131:e143380. doi: 10.1172/JCI143380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Kundu R, Narean JS, Wang L, Fenn J, Pillay T, Fernandez ND, Conibear E, Koycheva A, Davies M, Tolosa-Wright M, Hakki S, Varro R, McDermott E, Hammett S, Cutajar J, Thwaites RS, Parker E, Rosadas C, McClure M, Tedder R, Taylor GP, Dunning J, Lalvani A. 2022. Cross-reactive memory T cells associate with protection against SARS-CoV-2 infection in COVID-19 contacts. Nat Commun 13:80. doi: 10.1038/s41467-021-27674-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. 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: 10.1038/s41586-021-04186-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Moss P. 2022. The T cell immune response against SARS-CoV-2. Nat Immunol 23:186–193. doi: 10.1038/s41590-021-01122-w [DOI] [PubMed] [Google Scholar]
- 86. Niessl J, Sekine T, Lange J, Konya V, Forkel M, Maric J, Rao A, Mazzurana L, Kokkinou E, Weigel W, Llewellyn-Lacey S, Hodcroft EB, Karlsson AC, Fehrm J, Sundman J, Price DA, Mjösberg J, Friberg D, Buggert M. 2021. Identification of resident memory CD8+ T cells with functional specificity for SARS-CoV-2 in unexposed oropharyngeal lymphoid tissue. Sci Immunol 6:eabk0894. doi: 10.1126/sciimmunol.abk0894 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. 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: 10.1126/science.abh1823 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Xiang T, Wang J, Zheng X. 2022. The humoral and cellular immune evasion of SARS-CoV-2 Omicron and sub-lineages. Virol Sin 37:786–795. doi: 10.1016/j.virs.2022.11.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Müller TR, Gao Y, Wu J, Ribeiro O, Chen P, Bergman P, Blennow O, Hansson L, Mielke S, Nowak P, Vesterbacka J, Akber M, Söderdahl G, Smith CIE, Loré K, Chen MS, Ljungman P, Ingelman-Sundberg HM, Ljunggren H-G, Österborg A, Sette A, Grifoni A, Aleman S, Buggert M. 2024. Memory T cells effectively recognize the SARS-CoV-2 hypermutated BA.2.86 variant. Cell Host Microbe 32:156–161. doi: 10.1016/j.chom.2023.12.010 [DOI] [PubMed] [Google Scholar]
- 90. Viana R, Moyo S, Amoako DG, Tegally H, Scheepers C, Althaus CL, Anyaneji UJ, Bester PA, Boni MF, Chand M, et al. 2022. Rapid epidemic expansion of the SARS-CoV-2 Omicron variant in southern Africa. Nature 603:679–686. doi: 10.1038/s41586-022-04411-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. 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 603:488–492. doi: 10.1038/s41586-022-04460-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Liu J, Chandrashekar A, Sellers D, Barrett J, Jacob-Dolan C, Lifton M, McMahan K, Sciacca M, VanWyk H, Wu C, Yu J, Collier A-RY, Barouch DH. 2022. Vaccines elicit highly conserved cellular immunity to SARS-CoV-2 Omicron. Nature 603:493–496. doi: 10.1038/s41586-022-04465-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Li Z-RT, Zarnitsyna VI, Lowen AC, Weissman D, Koelle K, Kohlmeier JE, Antia R. 2019. Why are CD8 T cell epitopes of human influenza A virus conserved? J Virol 93. doi: 10.1128/JVI.01534-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Koutsakos M, Rowntree LC, Hensen L, Chua BY, Sandt CE, J H, W J, X K, L A, TM P, et al. 2021. Integrated immune dynamics define correlates of COVID-19 severity and antibody responses. Cell Rep Med:100208. doi: 10.2139/ssrn.3701261 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Du J, Wei L, Li G, Hua M, Sun Y, Wang D, Han K, Yan Y, Song C, Song R, Zhang H, Han J, Liu J, Kong Y. 2021. Persistent high percentage of HLA-DR+CD38high CD8+ T cells associated with immune disorder and disease severity of COVID-19. Front Immunol 12:735125. doi: 10.3389/fimmu.2021.735125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, Liu S, Zhao P, Liu H, Zhu L, Tai Y, Bai C, Gao T, Song J, Xia P, Dong J, Zhao J, Wang FS. 2020. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med 8:420–422. doi: 10.1016/S2213-2600(20)30076-X [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Bergamaschi L, Mescia F, Turner L, Hanson AL, Kotagiri P, Dunmore BJ, Ruffieux H, Sa A, Huhn O, Morgan MD, et al. 2021. Longitudinal analysis reveals that delayed bystander CD8+ T cell activation and early immune pathology distinguish severe COVID-19 from mild disease. Immunity 54:e8. doi: 10.1016/j.immuni.2021.05.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Vijayakumar B, Boustani K, Ogger PP, Papadaki A, Tonkin J, Orton CM, Ghai P, Suveizdyte K, Hewitt RJ, Desai SR, Devaraj A, Snelgrove RJ, Molyneaux PL, Garner JL, Peters JE, Shah PL, Lloyd CM, Harker JA. 2022. Immuno-proteomic profiling reveals aberrant immune cell regulation in the airways of individuals with ongoing post-COVID-19 respiratory disease. Immunity 55:542–556. doi: 10.1016/j.immuni.2022.01.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Georg P, Astaburuaga-García R, Bonaguro L, Brumhard S, Michalick L, Lippert LJ, Kostevc T, Gäbel C, Schneider M, Streitz M, et al. 2022. Complement activation induces excessive T cell cytotoxicity in severe COVID-19. Cell 185:493–512. doi: 10.1016/j.cell.2021.12.040 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Maurice NJ, Taber AK, Prlic M. 2021. The ugly duckling turned to swan: a change in perception of bystander-activated memory CD8 T cells. J Immunol 206:455–462. doi: 10.4049/jimmunol.2000937 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Kim TS, Shin EC. 2019. The activation of bystander CD8+ T cells and their roles in viral infection. Exp Mol Med 51:1–9. doi: 10.1038/s12276-019-0316-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Yin K, Peluso MJ, Luo X, Thomas R, Shin M-G, Neidleman J, Andrew A, Young KC, Ma T, Hoh R, et al. 2024. Long COVID manifests with T cell dysregulation, inflammation and an uncoordinated adaptive immune response to SARS-CoV-2. Nat Immunol 25:218–225. doi: 10.1038/s41590-023-01724-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. LaVergne SM, Dutt TS, McFann K, Baxter BA, Webb TL, Berry K, Tipton M, Stromberg S, Sullivan BM, Dunn J, Henao-Tamayo M, Ryan EP. 2023. Persistent CD8+ T cell proliferation and activation in COVID-19 adult survivors with post-acute sequelae: a longitudinal, observational cohort study of persistent symptoms and T cell markers. Front Immunol 14:1303971. doi: 10.3389/fimmu.2023.1303971 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Wiech M, Chroscicki P, Swatler J, Stepnik D, De Biasi S, Hampel M, Brewinska-Olchowik M, Maliszewska A, Sklinda K, Durlik M, Wierzba W, Cossarizza A, Piwocka K. 2022. Remodeling of T cell dynamics during long COVID is dependent on severity of SARS-CoV-2 infection. Front Immunol 13:886431. doi: 10.3389/fimmu.2022.886431 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Rowntree LC, Audsley J, Allen LF, McQuilten HA, Hagen RR, Chaurasia P, Petersen J, Littler DR, Tan HX, Murdiyarso L, et al. 2024. SARS-CoV-2-specific CD8(+) T cells from people with long COVID establish and maintain effector phenotype and key TCR signatures over 2 years. Proc Natl Acad Sci U S A 121:e2411428121. doi: 10.1073/pnas.2411428121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Klein J, Wood J, Jaycox JR, Dhodapkar RM, Lu P, Gehlhausen JR, Tabachnikova A, Greene K, Tabacof L, Malik AA, et al. 2023. Distinguishing features of long COVID identified through immune profiling. Nature 623:139–148. doi: 10.1038/s41586-023-06651-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Li J, Zaslavsky M, Su Y, Guo J, Sikora MJ, Unen V, Christophersen A, Chiou SH, Chen L, Li J, et al. 2022. KIR(+)CD8(+) T cells suppress pathogenic T cells and are active in autoimmune diseases and COVID-19. Science 376:eabi9591. doi: 10.4049/jimmunol.208.Supp.165.17 [DOI] [PMC free article] [PubMed] [Google Scholar]
