Next Article in Journal
A Pleomorphic Puzzle: Heterogeneous Pulmonary Vascular Occlusions in Patients with COVID-19
Next Article in Special Issue
Identifying SARS-CoV-2 Drugs Binding to the Spike Fatty Acid Binding Pocket Using In Silico Docking and Molecular Dynamics
Previous Article in Journal
Post-Translational Modification of ZEB Family Members in Cancer Progression
Previous Article in Special Issue
COVID-19 Molecular Pathophysiology: Acetylation of Repurposing Drugs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antigen-Specific T Cells and SARS-CoV-2 Infection: Current Approaches and Future Possibilities

1
Lambda Life a.s., Levocska 3617/3, 851 01 Bratislava, Slovakia
2
GAMMA-ZA s.r.o., Kollarova 8, 911 01 Trencin, Slovakia
3
Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc, University Hospital Olomouc, Hnevotinska 976/3, 77900 Olomouc, Czech Republic
4
Department of Laboratory Medicine, Faculty of Health Care and Social Work, Trnava University, Univerzitne Namestie 1, 918 43 Trnava, Slovakia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(23), 15122; https://doi.org/10.3390/ijms232315122
Submission received: 11 November 2022 / Revised: 25 November 2022 / Accepted: 26 November 2022 / Published: 1 December 2022

Abstract

:
COVID-19, a significant global health threat, appears to be an immune-related disease. Failure of effective immune responses in initial stages of infection may contribute to development of cytokine storm and systemic inflammation with organ damage, leading to poor clinical outcomes. Disease severity and the emergence of new SARS-CoV-2 variants highlight the need for new preventative and therapeutic strategies to protect the immunocompromised population. Available data indicate that these people may benefit from adoptive transfer of allogeneic SARS-CoV-2-specific T cells isolated from convalescent individuals. This review first provides an insight into the mechanism of cytokine storm development, as it is directly related to the exhaustion of T cell population, essential for viral clearance and long-term antiviral immunity. Next, we describe virus-specific T lymphocytes as a promising and efficient approach for the treatment and prevention of severe COVID-19. Furthermore, other potential cell-based therapies, including natural killer cells, regulatory T cells and mesenchymal stem cells are mentioned. Additionally, we discuss fast and effective ways of producing clinical-grade antigen-specific T cells which can be cryopreserved and serve as an effective “off-the-shelf” approach for rapid treatment of SARS-CoV-2 infection in case of sudden patient deterioration.

1. Introduction

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become one of the main causes of death worldwide, and the pandemic continues despite the development of highly efficacious vaccines. An important hallmark of severe infection is continued T cell depletion, resulting in uncontrolled inflammatory response with massive cytokine and chemokine release (so-called “cytokine storm”) that can progress to systemic inflammation, multi-organ damage and even death [1,2,3]. T cells and virus-specific T cells are essential to protect organisms against viral infection. Patients with a susceptible immune system may therefore be at great risk of fatal outcomes following SARS-CoV-2 infection. Indeed, elevated hospitalization and mortality rates have been described in immunocompromised patients [4,5,6,7]. Remy et al. [8] have recently suggested that immune depression, rather than immune hyperactivation, could be responsible for the clinical pathology of severe COVID-19, although both T cell hyperactivation and exhaustion have been reported in COVID-19 [9].
To date, there are no approved direct antiviral therapies to treat COVID-19 patients, and current curative strategies focus only on supportive care and treatment of disease symptoms (European Medicines Agency, EMA; U.S. Food and Drug Administration, FDA). Generally, many therapeutic options for viral diseases have severe adverse effects, could develop resistance or fail to ensure long-term protection. Therefore, adoptive cell therapy using virus-specific T cells (VSTs) has been used as an alternative treatment strategy, emerging as a promising approach due to its minimal toxicity [10,11,12,13]. Donor-derived T cells with specificity for adenovirus (AdV), cytomegalovirus (CMV), Epstein-Barr virus (EBV), BK virus or human herpesvirus have been used in previous studies to prevent or treat viral infections in the settings of immunity errors or after transplantation, demonstrating its safety and efficacy [10,12,14,15,16,17,18]. In addition to the reconstitution of antiviral immunity after treatment, it has been shown that VSTs persist for years after infusion and thus can provide long-term protection [19,20]. Data on SARS-CoV-1 infection have shown that T cell responses can last up to 17 years, while antibodies often weaken 1–2 years after disease [21]. The potential application of immunotherapy could also be relevant to patients at high risk of severe COVID-19, especially immunocompromised individuals with T cell deficiency or those receiving immunosuppressive therapies. This approach may decrease initial viral load [22], increase early bystander CD8+ T cell activation [23] and ensure adequate early type I interferon response [24,25], leading to enhanced viral clearance and prevention of severe disease development. Cytokine release syndrome, a major complication of COVID-19 [26], has been reported also with AdV, CMV or EBV infections [11,27,28], where VST therapy has been successfully used to treat many patients suffering from severe infections, with minimal complications [10,11,12,13]. Adoptively infused SARS-CoV-2-specific T cells (CSTs) are believed to target and kill virus-infected myeloid cells, resulting in breaking of the vicious cycle which drives the cytokine storm [29]. Infusion of CSTs could therefore be a life-saving strategy for severe SARS-CoV-2-infected patients.

2. Mechanism of Cytokine Storm Development in COVID-19

Once the virus enters the body, viral components are recognized by Toll-like receptors (TLRs) expressed on the surface of antigen-presenting cells (APCs), which rapidly induce anti-viral responses via the production of pro-inflammatory cytokines and type I interferons in order to clear infection [30]. Under normal conditions, innate (natural killer, NK) and adaptive (T cells) immune responses tend to cause an apoptosis of APCs and thus prevent unnecessary hyperactivation. However, any alterations in the lymphocyte activity caused by SARS-CoV-2 infection leads to the inactivation of both NK and cytotoxic T cells and their inability to kill APCs, resulting in dysregulation of immune balance, recruitment of more inflammatory cells, abnormal release of cytokines at the site of infection and induction of cytokine storm. Dysregulation of T cells induced by SARS-CoV-2 infection is described in more detail in the next chapter. The most commonly affected organ in COVID-19 patients are the lungs, manifesting as severe pneumonia, pulmonary edema and acute respiratory distress syndrome (ARDS) [31,32]. Subsequently, impaired vascular permeability due to cytokine storm further leads to cytokine release into the systemic circulation, damage of other organs and even death [3]. However, significant differences in clinical manifestation of COVID-19 between adults and children have been observed. SARS-CoV-2 infection of children is mostly asymptomatic or leads to a mild illness. Some pediatric patients can develop a post-infectious multisystem inflammatory syndrome in children (MIS-C), but these cases and mortality are rare. The immunological basis and immune mechanisms of differing disease severity and outcomes between children and adults have not been yet sufficiently characterized, but age-dependent factors and less comorbidities have been suggested to modulate antiviral immune response. Nonetheless, this issue is beyond the scope of this review and was described in more detail by others [33,34,35,36].
Viral components are recognized by Toll-like receptors (TLRs). It has been suggested that cell surface TLR4 and intracellular TLR3, TLR7 and TLR8 contribute to antiviral responses against SARS-CoV-2 infection (Figure 1) [37,38]. TLR4 recognizes and binds SARS-CoV-2 spike (S) glycoprotein [39,40], TLR7/8 detects viral single-stranded ribonucleic acid (ssRNA) and ligand for TLR3 is a viral double-stranded RNA ribonucleic acid (dsRNA) [38,41]. TLRs trigger two signal transduction pathways, (i) the myeloid differentiation primary response protein 88 (MyD88)-dependent with MyD88 adaptor-like protein (MAL), leading to activation of nuclear factor kappa B (NF-κB) pathway and rapid production of pro-inflammatory cytokines such as interleukin (IL)-6, IL-1β or tumor necrosis factor α (TNF-α), and (ii) the MyD88-independent pathway mediated through Toll/interleukin-1 receptor (TIR)-domain-containing adaptor protein (TRIF) and TRIF-related adaptor molecule (TRAM), leading to release of type I interferons (IFN-I; IFN-α/β) through the induction of interferon regulatory factor-3 (IRF-3), and late NF-κB activation. TLR4 mediates signals through both pathways, TLR3 through TRIF and TLR7/8 utilize the MyD88 pathway. Both pathways contribute to elevated levels of cytokines in the body [3,38,42,43]. IFN-I response is critical to combat viral infections. However, it is quickly and selectively abrogated by SARS-CoV-2 via different mechanisms [44,45]. This inhibition ability is associated with clinical severity of the disease [46]. Clinical studies have demonstrated that coronaviruses are able to evade innate immunity during the first 10 days of infection, allowing elevated virus replication, which corresponds to a period of widespread inflammation and steadily increasing viral load [47,48]. Indeed, several groups have reported remarkably reduced IFN-I response in severe COVID-19 patients [24,25,49]. This is accompanied by an overwhelming production of pro-inflammatory cytokines, accumulation of pathogenic monocyte-macrophages, and suboptimal T cell response, leading to hyperinflammation and cytokine storm [44,50]. In addition to the direct role of TLR4 signaling in mediating the immunopathogenesis, TLR4 activation resulting from its interaction with spike (S) protein also increases cell surface expression of key SARS-CoV-2 receptor angiotensin-converting enzyme 2 (ACE2), which in turn augments viral load as well as hyperinflammatory events [51].
The interaction between ACE2 and S protein of SARS-CoV-2 is responsible for viral entry to the host cells (Figure 1) [52,53,54,55]. Additionally, S protein cleavage by cellular transmembrane serine protease 2 (TMPRSS2) is required to allow fusion of viral and cellular membranes [52,55]. In the respiratory tract, ACE2 and TMPRSS2 are co-expressed in alveolar epithelial type II (ATII) cells [56], alveolar macrophages [54,57,58] and nasal goblet secretory cells [56]. Another enzyme involved in the viral entry process is furin, which is highly expressed in lung tissue [54,59,60]. ACE2, TMPRSS2 and furin triple expression has been found in lung macrophages [57]. If ACE2-virus complex could not find TMPRSS2 or the target cell does not express enough TMPRSS2, the virus is internalized into the endolysosomes, where it is cleaved by cathepsins (Figure 1). After the fusion of viral and cellular membranes, viral ribonucleic acid (RNA) is released into the cytoplasm and can be replicated into dsRNA [55]. ssRNA and dsRNA are then recognized by TLR7/8 and TLR3, respectively, and trigger pro-inflammatory pathways as mentioned above [38,41].
ACE2 has been described as an important cell-protective component as it acts like a negative regulator of the renin-angiotensin pathway (RAS), thus having an anti-inflammatory and anti-fibrotic effect. SARS-CoV-2 triggers ACE2 downregulation via its cleavage by TMPRSS2 and A disintegrin and a metalloproteinase 17 (ADAM17) enzyme, leading to imbalance between ACE2 and its counterpart ACE [61,62,63,64]. ACE upregulates the level of angiotensin II (Ang2), which excessively binds to angiotensin II type-1 receptor (AT1R) and triggers inflammatory response. Ang2 interacts with both tissue-resident and immune cells. Activated tissue-resident cells produce pro-inflammatory factors, such as NF-κB, TNF-α, IL-6, IL-1β, IFN-γ, prostaglandins and vascular endothelial cell growth factor (VEGF). Moreover, Ang2 promotes immune cell recruitment into the site of injury and stimulates the production of cytokines and chemokines, resulting in amplification of inflammatory cycle and tissue damage (Figure 2) [62,65,66]. Additionally, Ang2 upregulates the expression of TLR4, leading to stimulation of NF-κB and induction of pro-inflammatory cytokines [67]. Highly elevated levels of cytokines, especially IL-6 increase vascular permeability, leading to release of pro-inflammatory cytokines to the systemic circulation, which further contribute to the cytokine storm and subsequent multi-organ failure [26,66].
Compared to adults, the lower expression of ACE2 has been described in nasal and bronchial tissue of children [68,69]. Also, other research groups have suggested that ACE2 expression in nasal and lung epithelium increases with age, which could be one of many possible explanations for the difference in COVID-19 severity observed between children and adults [70,71]. Moreover, expression of TMPRSS2 rises with age, and is significantly higher in the airway epithelium of adults compared to infants [34,71,72].

3. Dysregulation of T Cells in COVID-19

T cells have a key role in immune reactions during COVID-19. CD4+ T helper cells regulate the development of high-affinity neutralizing antibodies, B cell differentiation into memory and long-lived antibody secreting cells and also CD8+ T cell activation through provision of soluble mediators and co-stimulation. CD8+ cytotoxic T lymphocytes secrete antiviral cytokines, such as IFN-γ and TNF-α, and directly kill virus-infected cells [9,73]. During a primary immune response, APCs identify and present foreign antigens to T cells, which recognize them through their T cell receptor (TCR) [74,75]. Once viral particles are recognized, the TCR signaling pathway triggers T cell activation, proliferation and differentiation into effector subsets. Initially, the expression of CD25 (cell surface marker of IL-2 receptor α-chain) [76,77] and level of IL-2 is rapidly upregulated in early activated T cells [78,79], creating a positive feedback loop for T cell activation and acquiring effector functions in order to suppress infection. The subsequent fate of activated cells differs in mild and severe SARS-CoV-2 infection (Figure 3). In mild COVID-19 patients, IL-2 drives differentiation into regulatory T cells (Tregs) expressing transcription factor forkhead box-p3 (FOXP3), which is crucial for maintaining immune tolerance and immune system homeostasis [80]. Simultaneously, the expression of immune checkpoint molecules, such as cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4) is increased, providing a negative feedback regulation. This leads to conversion of early activated T cells into the suppressive state and resolution of inflammation [81,82]. Following antigen clearance, most activated effector T cells die by apoptosis, but a subset persists and differentiate into long lived-memory cells [83]. In severe COVID-19 patients, T cell expression of FOXP3 is repressed, likely due to high levels of pro-inflammatory cytokines, especially IL-6, which has emerged as a key inducer of the cytokine storm [84,85]. Therefore, highly activated effector T cells continuously produce multiple pro-inflammatory cytokines and furin [79,86,87]. Furin can further enhance SARS-CoV-2 entry to host cells, as mentioned above. Prolonged immune activation alters the lymphocyte secretome. Activated T cells are differentiated into a CD25+ IL-10R+ phenotype, resulting in production of IL-10 rather than IL-2 [88]. Under these conditions, the IL-10 regulatory loop is created and amplified, resulting in loss of CD4+ and CD8+ cell functions and inability to respond against pathogens efficiently [89,90,91,92]. A recent study showed that dramatic early IL-10 elevation may contribute to COVID-19 severity by promoting T cell exhaustion [93]. Furthermore, inhibitory molecules are overexpressed. Programmed cell death 1 receptor (PD-1) [1,83,94,95], CTLA-4 [95,96], T cell immunoglobulin and mucin-domain containing-3 (Tim-3) [1,97], lymphocyte-activation gene 3 (LAG-3) [95], B- and T-lymphocyte attenuator (BTLA) [98] and inhibitory receptor NKG2A [99] have been identified as markers of chronic activation, inhibition and exhaustion in both CD4+ and CD8+ T cells. Because of T cell inability to clear pathogen, viral load and antigen exposure time increase, further contributing to exhaustion [100]. Several studies have claimed that CD8+ T cells primed during chronic infections are able to recover and differentiate into memory T cells after adoptive transfer into uninfected mice in the course of the first one to three weeks of infection, while longer exposure to infection results in irreversible exhaustion [101,102,103]. It has been shown that frequency of multi-functional CD4+ T cells and non-exhausted CD8+ T cells were significantly lower in severe COVID-19 cases compared with mild group and healthy controls, whereas the number of non-functional subsets significantly increased [1,104]. Hence, SARS-CoV-2 infection may break down antiviral immunity at an early stage of disease. However, the exact mechanism is not fully understood yet. Moreover, SARS-CoV-2 can directly infect T cells via ACE2 receptor expressed on T cells [105], which results in T cell death and potentially contributes to reduced T cell number and immune dysregulation observed in COVID-19 illness. Several studies have claimed that severe SARS-CoV-2 infection is associated with marked reduction of all lymphocyte subsets (lymphopenia) that correlate with morbidity and mortality [1,94,106,107]. Therefore, immune reconstitution, marked by increased T cell number, is crucial for COVID-19 patients.
The situation is slightly different in pediatric patients. Studies have shown that only 3.5–5.2% of children with COVID-19 had lymphopenia, and the majority had normal leukocyte numbers [33,108,109]. Furthermore, differences in interferon signaling have been investigated in children and adults with COVID-19. Children can develop more robust interferon response after TLR stimulation [35,110]. It has been reported that increased production of IFN-γ during an early immune response can result in a more rapid clearance of viral infection [33]. It has been also shown that SARS-CoV-2-specific T cell responses differ in infected adults and children. Acute and memory CD4+ T cell reactivity to structural SARS-CoV-2 proteins increase with age, whereas CD8+ T cell responses increase with time post-infection. Moreover, CD4+ T cells are targeting primarily structural proteins in adults, while pediatric CD4+ T cells significantly target non-structural proteins in open reading frame (ORF) 1ab [34]. Some of them are able to block the host interferon response [111], which could contribute to the age-associated decline of IFN response to infection by SARS-CoV-2. Likewise, children’s adaptive immune system differs from adults. Thus, the immune system of pediatric patients is less likely to overreact to SARS-CoV-2 infection. Stronger T-cell response to SARS-CoV-2 spike protein and a higher neutralizing antibody count were observed in adults compared to children [33]. Taken together, milder adaptive immune response, but more robust innate response in children may prevent a cytokine storm, possibly eliminating an unwanted overreaction to the virus. Despite that, immunological differences with adults are not fully understood, and further research is still needed to discern the protective role of T cells in children with COVID-19.

4. SARS-CoV-2-Specific T Cell Therapy for the Treatment and Prevention of Severe COVID-19 Infection

Several recent studies have pointed out a link between severe COVID-19 and insufficient early innate immune responses to SARS-CoV-2, leading to weak or delayed adaptive response [107,112]. In primary (SARS-CoV) infection, CD4+ T cells settle virus control and affect disease progression. Aged BALB/c mice with CD4+ T cell depletion experienced upregulated cytokine production, protracted SARS-CoV clearance from the lungs, decline in neutralizing antibody titers and pulmonary influx of lymphocytes [113]. Nucleocapsid (N)-specific airway memory CD4+ T cells produced via intranasal vaccination successfully protected against lethal disease via accelerated production of IFN-γ. Airway depletion of CD4+ cells and IFN-γ lowered survival rate and manifested as elevated pathological changes in the lungs [114]. Similar results were also described in other animal models [115,116,117]. Furthermore, murine studies of pathogenic coronavirus infection largely demonstrated a protective role for T cells and suggested that SARS CoV-2-specific CD4+ and CD8+ T cells will control virus replication and moderate the pathology associated with COVID-19 [118].
Analysis of bronchoalveolar lavage fluid (BALF) from COVID-19 patients revealed decreased clonal T cell expansion and suboptimal presence of CSTs in the lungs of those with severe or fatal disease [119,120]. Moreover, depletion of CD8+ T cells in hematologic malignancy patients with impaired humoral responses to SARS-CoV-2 has been associated with poor outcomes [121]. Indeed, lymphopenia is a typical profile in patients with COVID-19, especially in severe cases [9]. It has been shown that the numbers of total T cells were negatively correlated to serum level concentrations of IL-6, IL-10, and TNF-α [1]. Thus, enhancing immune reconstitution, especially CSTs, is the most crucial process which takes place in treatment of COVID-19 patients. In a convalescent patient cohort, CD4+ and CD8+ CSTs were detected in peripheral blood mononuclear cells (PBMCs) from more than 90% and 70% of individuals, respectively. Presence of CSTs positively corelated with recovery and decreased illness severity [73,122,123,124,125]. These findings indicate the importance of T cell responses, particularly CSTs, for effective viral clearance in patients with acute infection. Studies have shown suboptimal responses of immunocompromised patients to vaccination [126,127,128,129,130,131]. Booster vaccines are required in this population, but it is unclear how durable will be their protective immunity compared with immunocompetent individuals. Adoptive cellular therapy with CSTs could therefore be a promising treatment or prophylactic strategy leading to improved clinical outcomes in patients with or at high risk for COVID-19. A review of VSTs given to 36 patients with primary immunodeficiency disorders reported excellent responses to VSTs both for prophylaxis (81% of patients protected from viral reactivation) and treatment (response rates 76–100% depending on the virus) [132]. Considering long-lasting T cell responses against SARS-CoV-1 [124,133] and Middle East respiratory syndrome coronavirus (MERS-CoV) [134], specific SARS-CoV-2 memory T cells are believed to sustain for a longer period with the ability to clear viral re-infection. Very recently, it was reported that T cell memory persists for 8 [123] to 10 months [135] after primary SARS-CoV-2 infection, which further supports the potential of CSTs to maintain durable protection against severe COVID-19 despite humoral immunity declines [136]. Several clinical trials are ongoing to determine the effect of CSTs for the treatment or prevention of SARS-CoV-2 infection (Table 1).

5. Large-Scale Production of SARS-CoV-2-Specific T-Cells

Production of VSTs for clinical use consists of the selection of potential donor, verification of his specific T cell frequencies and prediction of T cell enrichment efficiency, subsequent VSTs ex vivo expansion from donor’s PBMCs and final quality control. T cell donor selection is based on human leukocyte antigen (HLA) type, viral serostatus and presence of VSTs in peripheral blood. It has been reported that SARS-CoV-2-specific CD4+ and CD8+ T cells are capable of activating and proliferating ex vivo for at least 10 months after infection [135]. Direct selection of VSTs uses major histocompatibility complex (MHC)–antigen multimers or cytokine capture system (CCS; IFN-γ capture system) after cell restimulation with viral antigens [137,138,139]. While the knowledge of immunodominant HLA-restricted peptides is required for MHC multimer technology [137,140], CCS does not have any HLA restrictions and can be performed with recombinant proteins or overlapping peptide pools spanning the entire viral proteome, enabling the generation of CD4+ and CD8+ T cells specific to multiple epitopes [138,141,142]. Antigen-specific T cells prepared by the IFN-γ CCS have been classified as an advanced-therapy medicinal product by the Committee for Advanced Therapies of the European Medicines Agency [143]. Briefly, suitable donor VSTs isolated from PBMCs are cultivated in the presence of viral antigens and growth-enhancing cytokines, such as IL-4 + IL-7 [15], IL-1 + IL-6 + IL-23 [144], IL-2 + IL-4 + IL-7 or IL-2 + IL-7 + IL-15 [29] for 10 to 12 days [139]. Such combinations of cytokines can promote the survival of activated T cells by upregulation of anti-apoptotic molecules and support the retention of their central memory phenotype [15,144]. Subsequently, IFN-γ+ virus-specific T cells are captured by magnetic cell sorting enrichment processes [138]. Several research groups have successfully generated CSTs using CliniMACS CCS [145,146,147,148,149].
Optimal HLA matching strategy is still under study. In the case of allogeneic hematopoietic stem cell transplantation (HSCT), seropositive donors can usually also provide T cell donation. However, some of them may not be available or have an insufficient amount of memory T cells in their peripheral blood despite seropositivity. Moreover, granulocyte colony-stimulating factor (G-CSF) treatment usually used for mobilization of PBMCs in healthy volunteers before stem cell donation has been shown to have long-term negative effects on T cell functional activity [150,151,152]. In general, adoptive transfer of donor-derived VSTs is highly patient-specific and requires virus-immune donors, which has emerged as a barrier that hinders broad implementation of this therapy. Thus, partially HLA-matched VSTs have been investigated as an alternative therapeutic option [13,153,154,155,156]. Such products can be cryopreserved and stored, allowing for rapid treatment of infection if necessary. Indeed, the safety and efficacy of this approach has been determined in several studies [19,154,157,158,159] and was shown to not induce de novo graft-versus-host disease (GvHD) despite in vitro recognition of recipient HLA molecules [157]. Available data indicate that at least one shared HLA allele is necessary to mediate antiviral T cell immunity [160]. Leung and colleagues [145] successfully prepared CSTs from six convalescent COVID-19 patients using IFN-γ CCS and SARS-CoV-2-specific PepTivators (peptide pools consisting mainly of 15-mer sequences with 11 amino acids overlap). They hypothesized 88 to 99% probability that the recipient will share at least one HLA allele with one of the six donors, depending on ethnic background. The peptide pool used in this study consists of 88 peptides with a length of 9–22 amino acids originating from structural and non-structural proteins. Sixty-three of these peptides are restricted to HLA class I, and 25 peptides are restricted to HLA class II. Generally, the selected peptides allow a broad HLA coverage throughout the Caucasian population and stimulate both CD4+ and CD8+ T cell subpopulations. HLA-A, B and DR alleles represented in this peptide pool are indeed present in >95%, >85% and >90% of the donors in the alloCELL registry (http://www.alloCELL.org (accessed on 9 September 2022)). Moreover, the majority of these peptides are identical in wild-type and presently circulating variants of concern, including Delta and Omicron. It is assumed that future emerging variants will also be adequately covered by used PepTivator SARS-CoV-2 Select, making it suitable for donor selection and subsequent clinical-scale manufacturing of CSTs [145,149]. Interestingly, Tzannou et al. [161] successfully prepared a small bank of VSTs from strategically selected third-party donors based on their HLA profile and immunity to the target of interest and thus provided potential treatment to a large racially and ethnically diverse patient population. The effect of partially HLA-matched CSTs in high-risk patients hospitalized with COVID-19 was described in clinical trial NCT04401410. One of them developed cytokine release syndrome symptoms but recovered, and the infection was successfully cleared in the remaining three patients after CST infusion [162]. However, because of the missing control group, the extent of the CST therapy cannot be clearly defined, and this trial was terminated due to insufficient number of recruited patients. Thus, the use of third-party donors together with quick and effective isolation methods (IFN-γ capture assay, MHC multimer technology) could be a promising approach to supply antigen-specific effector cells for treatment of potentially life-threatening infections in immunocompromised patients.
CST products can be generated not only from virus-experienced but also virus-naive donors, as SARS-CoV-2-specific T cell responses were observed in some unexposed individuals, likely due to pre-existing cross-reactivity with common seasonal coronaviruses [73,146,163,164,165]. The pool of potential donors can be also enriched with individuals vaccinated against SARS-CoV-2 [166,167,168], whose specific T cell responses have been proven effective against other virus variants [169,170]. The generation of CSTs from vaccinated donors on a clinically relevant scale was successfully demonstrated for the first time by Taborska et al. [171]. Such cross-reactivity of generated CSTs against new viral strains is essential for its therapeutic use. It has been suggested that most of the memory CD4+ T cell response is conserved against SARS-CoV-2 variants of concern [170]. Indeed, the vast majority of T cell epitopes identified in recovering patient CSTs appeared to be conserved in SARS-CoV-2 genome modifications [15,172,173,174,175]. The strong cross-reactivity of CSTs against circulating variants has been detected in several studies [166,168,169,176]. Moreover, CSTs from convalescent patients were able to respond to viral strains partially resistant to vaccine-induced humoral immunity [177,178,179]. However, other research groups analyzed the response of CD8+ T cells and have found that the ability of some mutant epitopes to bind to certain HLAs is decreased [180,181]. Thus, it is important to ensure that CSTs manufactured for clinical practice are effective against new emerging SARS-CoV-2 variants.
Because of the hyperinflammatory nature of COVID-19, many patients with severe disease receive a systemic corticosteroid therapy, which may improve clinical outcomes [182,183]. However, commonly used dexamethasone is highly lymphocytotoxic [184,185]. This could significantly limit the efficacy of CST treatment as corticosteroids are highly likely to induce apoptosis of adoptively transferred T cells. Therefore, the question arises as to how effective T-cell therapy would be in the setting of pneumonia or ARDS. Very recently, Basar et al. [29] proposed an efficient strategy to inactivate the glucocorticoid receptor gene (NR3C1) in cytotoxic T lymphocytes using CRISPR-Cas9 gene editing. Such modified cells retained viability and effector functions in the presence of dexamethasone in vitro. This approach could make CST application clinically feasible even when simultaneous immunosuppressive pharmacotherapy is required.

6. Other Potential Cell-Based Therapies for COVID-19

In addition to antigen-specific T cells, therapies using NK cells, Tregs or mesenchymal stem cells (MSCs) could potentially have a beneficial effect for COVID-19 patients. NK cells are an essential part of antiviral defense, as evidenced by inborn errors of NK cell development that led to increased viral susceptibility [186,187]. NK cell exhaustion and their low numbers in the blood are common in patients with moderate and severe COVID-19 disease [188,189,190], which may result in decreased clearance of infected and activated cells, and elevation of inflammation markers. Indeed, inverse correlations between NK cell counts and serum levels of IL-6 [189] and interleukin-2 receptor alpha [191] have been reported. The potential role of NK cells in fighting viral infections, including COVID-19, has been mentioned in several publications [192,193,194,195]. It turns out that adoptive transfer of highly activated NK cells at the beginning of the disease might help to boost innate and adaptive immunity, improving survival and reducing disease progression rates in COVID-19 patients [196]. Furthermore, Ma et al. [197] proved that NK cells modified with a chimeric antigen receptor (CAR) using single-chain variable fragment (scFv) domain of S309, a monoclonal antibody targeting SARS-CoV-2 S glycoprotein, successfully killed spike-expressing target cells and were also cross-reactive with other spike variants. There are several clinical trials evaluating safety and efficiency of immunotherapy with allogeneic NK cells as an adjunctive treatment for SARS-CoV-2 infection (NCT04900454, NCT04280224, NCT04365101, NCT04634370, NCT04363346, NCT04578210) and one trial focused on CAR-modified NK cells targeting the S protein of SARS-CoV-2 (NCT04324996).
The therapeutic potential of ex vivo expanded MSCs and Tregs in ARDS is based on their immunomodulatory and tissue reparative properties. Promising results of both treatments have been shown in preclinical and early clinical trials [198,199,200,201,202,203]. MSCs are unlikely to be infected by SARS-CoV-2 as they do not express ACE2 and TMPRSS2 receptors [204,205,206]. As a result, they may be used for treatment of affected tissues without being destroyed by the virus. In addition to regenerative potency [207], MSCs are able to inhibit the overactivated immune system [208,209] and also repair defective immune function [210]. Importantly, they can suppress cytokine production and mitigate cytokine storm [204,211,212]. Avoiding the cytokine storm may be the key for the treatment of COVID-19 patients. Beneficial properties of MSCs are mediated by direct interaction with immune cells and secretion of paracrine factors, such as keratinocyte growth factor, hepatocyte growth factor or angiopoietin-1, which have been linked with lung protection [204,213,214,215]. The advantage of MSCs is their low surface expression of HLA antigens, which allows for allogeneic infusion without HLA matching [216,217,218]. MSCs were used in several pilot studies for treatment of severe COVID-19, demonstrating their efficiency by patient clinical improvement, better oxygenation and decreased inflammatory markers (CRP, IL-6, TNF-α, procalcitonin) with no observed adverse effects [204,219,220,221,222]. Promising preliminary results of MSC efficacy were also shown in randomized controlled trials [203,223,224,225]. Interestingly, MSC-derived exosomes appear to be a promising therapeutic alternative for severe COVID-19 as they are easy to produce and supply, having comparable therapeutic efficacy to MSCs administration [226,227].
It has been shown that Tregs are able to hinder activation of innate as well as adaptive immune cells by inhibitory surface molecules (LAG-3; CTLA-4) and secretion of immunosuppressive cytokines (IL-10, IL-35, transforming growth factor beta-TGF-β), which points to their importance in prevention of inflammation-induced tissue damage during acute infections and tissue reparation [200,228,229,230]. Peripheral Treg count is prominently reduced in severely ill COVID-19 patients [231,232,233,234], which might be one of the reasons for the immune system hyperactivation (massive proliferation and activation of macrophages, neutrophils, dendritic cells, mast cells and Th17 cells) and lung damage [235]. Notably, depletion of Tregs from mice infected with murine coronavirus led to increased mortality with acute encephalitis, underlining the protective nature of Tregs during acute coronavirus infection [236]. Recently, two patients with ARDS mediated by COVID-19 were treated with ex vivo expanded allogeneic Tregs, resulting in symptom improvement, rapid decrease in inflammatory markers (IL-6, IL-12, TNF-α, IFN-γ) and no adverse effects related to the treatment [237]. Treatment of SARS-CoV-2-induced ARDS by allogeneic ex vivo expanded Tregs is evaluated in clinical trials NCT04468971 and NCT05027815.

7. Conclusions

In this review, we discussed the pathology of SARS-CoV-2 infection, main pitfalls and therapeutic strategies and also the numerous data highlighting the important role of CD4+ and CD8+ virus-specific T cells during COVID-19. We have also considered quantitative aspects and outlined possible treatment approaches which might still have some limitations. The cellular part of immune response against SARS-CoV-2 is characterized mainly by antiviral T cells with a broad repertoire, being detectable for more than 1 year after initial infection. In contrast to antibody generation, the protective effect of T cell response is largely also maintained toward viral variants of concern. As T cells play critical roles in the clearance of SARS-CoV-2 and protection from development of severe COVID-19, immune suppressed individuals have a higher risk for severe SARS-CoV-2 infection. This population as well as COVID-19 patients with inadequate endogenous T cell responses may benefit from adoptive transfer of allogeneic, SARS-CoV-2-specific T cells isolated from convalescent individuals. Adoptive T cell therapy is being explored as a preventative or therapeutic adjunctive therapy against SARS-CoV-2, with very promising results as enriched SARS-CoV-2-specific T cells seems to maintain proliferative capacity and cytotoxic potential toward target cells. Therefore, enriching antiviral memory T cells and depletion of potentially alloreactive naive T cells alongside precise consideration of partial HLA class matching between donor and recipient, with subsequent adoptive transfer of SARS-CoV-2-specific T cells should provide an exceptional and safe treatment option for COVID-19 patients at risk of severe disease. Infusion of SARS-CoV-2-specific T cells could be a life-saving strategy for severe SARS-CoV-2-infected patients.

Author Contributions

Conceptualization, Z.N.; writing—original draft preparation Z.N.; writing—review and editing, Z.N., T.Z. and N.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Operational Program Integrated Infrastructure for the project: Development and implementation of immunomodulating cell therapy in the fight against COVID-19 pandemic (ITMS2014+: 313011ATT8), co-financed from EU sources.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADAM17 A disintegrin and metalloproteinase 1
AdVadenovirus
Angangiotensin
APCantigen-presenting cell
ARDSacute respiratory distress syndrome
ATIIalveolar epithelial type II
AT1Rangiotensin II receptor type 1
BALFbronchoalveolar lavage fluid
BTLAB- and T-lymphocyte attenuator
CARchimeric antigen receptor
CCScytokine capture system
CD25cluster of differentiation 25
CD4cluster of differentiation 4
CD8cluster of differentiation 8
CMVcytomegalovirus
COVID-19coronavirus disease 2019
CSTSARS-CoV-2-specific T cell
CTLA-4cytotoxic T-lymphocyte–associated antigen 4
dsRNAdouble-stranded RNA
EBVEpstein-Barr virus
EMAEuropean Medicines Agency
FDAThe United States Food and Drug Administration
FOXP3forkhead box-p3
G-CSFgranulocyte colony-stimulating factor
GvHDgraft-versus-host disease
HLA human leukocyte antigen
HSCThematopoietic stem cell transplantation
IFNinterferon
ILinterleukin
IL-2Rinterleukin 2 receptor
IL-10Rinterleukin 10 receptor
IRFinterferon regulatory factor
LAG-3lymphocyte-activation gene 3
MALMyD88 adaptor-like protein
MasRMas receptor
MERS-CoV Middle East respiratory syndrome coronavirus
MHCmajor histocompatibility complex
MIS-Cmultisystem inflammatory syndrome in children
MSCmesenchymal stem cell
MyD88myeloid differentiation primary response protein 88
N proteinnucleocapsid protein
NF-κBnuclear factor kappa B
NK cell natural killer cell
NKG2ACD94/NK group 2 member A
ORF open reading frame
PBMCsperipheral blood mononuclear cells
PD1programmed cell death 1 receptor
RAS renin–angiotensin system
RNAribonucleic acid
S protein spike protein
SARS-CoVsevere acute respiratory syndrome coronavirus
SARS-CoV-1severe acute respiratory syndrome coronavirus 1
SARS-CoV-2severe acute respiratory syndrome coronavirus 2
scFvsingle chain variable fragment
ssRNAsingle-stranded ribonucleic acid
TCRT cell receptor
TGF-βtransforming growth factor beta
Tim-3T cell immunoglobulin and mucin-domain containing-3
TIRToll/interleukin receptor
TLRToll-like receptor
TMPRSS2transmembrane serine protease 2
TNF-αtumor necrosis factor α
TRAMTRIF-related adaptor molecule
Tregregulatory T cell
TRIF TIR domain-containing adaptor-inducing interferon-β
VSTvirus-specific T cell

References

  1. Diao, B.; Wang, C.; Tan, Y.; Chen, X.; Liu, Y.; Ning, L.; Chen, L.; Li, M.; Liu, Y.; Wang, G.; et al. Reduction and Functional Exhaustion of T Cells in Patients with Coronavirus Disease 2019 (COVID-19). Front. Immunol. 2020, 11, 827. [Google Scholar] [CrossRef] [PubMed]
  2. Toor, S.M.; Saleh, R.; Nair, V.S.; Taha, R.Z.; Elkord, E. T-cell responses and therapies against SARS-CoV-2 infection. Immunology 2020, 162, 30–43. [Google Scholar] [CrossRef] [PubMed]
  3. Manik, M.; Singh, R.K. Role of toll-like receptors in modulation of cytokine storm signaling in SARS-CoV-2-induced COVID-19. J. Med. Virol. 2021, 94, 869–877. [Google Scholar] [CrossRef] [PubMed]
  4. Deng, G.; Yin, M.; Chen, X.; Zeng, F. Clinical determinants for fatality of 44,672 patients with COVID-19. Crit. Care 2020, 24, 179. [Google Scholar] [CrossRef]
  5. Kuderer, N.M.; Choueiri, T.K.; Shah, D.P.; Shyr, Y.; Rubinstein, S.M.; Rivera, D.R.; Shete, S.; Hsu, C.-Y.; Desai, A.; de Lima Lopes, G., Jr.; et al. Clinical impact of COVID-19 on patients with cancer (CCC19): A cohort study. Lancet 2020, 395, 1907–1918. [Google Scholar] [CrossRef]
  6. Dai, M.; Liu, D.; Liu, M.; Zhou, F.; Li, G.; Chen, Z.; Zhang, Z.; You, H.; Wu, M.; Zheng, Q.; et al. Patients with cancer appear more vulnerable to SARS-COV-2: A multicenter study during the COVID-19 outbreak. Cancer Discov. 2020, 10, 783–791. [Google Scholar] [CrossRef]
  7. Sharma, A.; Bhatt, N.S.; Martin, A.S.; Abid, M.B.; Bloomquist, J.; Chemaly, R.F.; Dandoy, C.; Gauthier, J.; Gowda, L.; Perales, M.-A.; et al. Clinical characteristics and outcomes of COVID-19 in haematopoietic stem-cell transplantation recipients: An observational cohort study. Lancet Haematol. 2021, 8, e185–e193. [Google Scholar] [CrossRef]
  8. Remy, K.E.; Mazer, M.; Striker, D.A.; Ellebedy, A.H.; Walton, A.H.; Unsinger, J.; Blood, T.M.; Mudd, P.A.; Yi, D.J.; Mannion, D.A.; et al. Severe immunosuppression and not a cytokine storm characterizes COVID-19 infections. J. Clin. Investig. 2020, 5, e140329. [Google Scholar] [CrossRef]
  9. DiPiazza, A.T.; Graham, B.S.; Ruckwardt, T.J. T cell immunity to SARS-CoV-2 following natural infection and vaccination. Biochem. Biophys. Res. Commun. 2020, 538, 211–217. [Google Scholar] [CrossRef]
  10. Bollard, C.M.; Heslop, H.E. T cells for viral infections after allogeneic hematopoietic stem cell transplant. Blood 2016, 127, 3331–3340. [Google Scholar] [CrossRef]
  11. McLaughlin, L.P.; Bollard, C.M.; Keller, M.D. Adoptive T Cell Therapy for Epstein–Barr Virus Complications in Patients With Primary Immunodeficiency Disorders. Front. Immunol. 2018, 9, 556. [Google Scholar] [CrossRef] [PubMed]
  12. Muftuoglu, M.; Olson, A.; Marin, D.; Ahmed, S.; Mulanovich, V.; Tummala, S.; Chi, T.L.; Ferrajoli, A.; Kaur, I.; Li, L.; et al. Allogeneic BK Virus–Specific T Cells for Progressive Multifocal Leukoencephalopathy. N. Engl. J. Med. 2018, 379, 1443–1451. [Google Scholar] [CrossRef] [PubMed]
  13. Tzannou, I.; Papadopoulou, A.; Naik, S.; Leung, K.; Martinez, C.A.; Ramos, C.A.; Carrum, G.; Sasa, G.; Lulla, P.; Watanabe, A.; et al. Off-the-Shelf Virus-Specific T Cells to Treat BK Virus, Human Herpesvirus 6, Cytomegalovirus, Epstein-Barr Virus, and Adenovirus Infections After Allogeneic Hematopoietic Stem-Cell Transplantation. J. Clin. Oncol. 2017, 35, 3547–3557. [Google Scholar] [CrossRef] [PubMed]
  14. Leen, A.M.; Christin, A.; Myers, G.D.; Liu, H.; Cruz, C.R.; Hanley, P.J.; Kennedy-Nasser, A.A.; Leung, K.S.; Gee, A.P.; Krance, R.A.; et al. Cytotoxic T lymphocyte therapy with donor T cells prevents and treats adenovirus and Epstein-Barr virus infections after haploidentical and matched unrelated stem cell transplantation. Blood 2009, 114, 4283–4292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Gerdemann, U.; Keirnan, J.M.; Katari, U.L.; Yanagisawa, R.; Christin, A.S.; Huye, L.E.; Perna, S.K.; Ennamuri, S.; Gottschalk, S.; Brenner, M.K.; et al. Rapidly Generated Multivirus-specific Cytotoxic T Lymphocytes for the Prophylaxis and Treatment of Viral Infections. Mol. Ther. 2012, 20, 1622–1632. [Google Scholar] [CrossRef] [Green Version]
  16. Gerdemann, U.; Katari, U.L.; Papadopoulou, A.; Keirnan, J.M.; Craddock, J.A.; Liu, H.; Martinez, C.A.; Kennedy-Nasser, A.; Leung, K.S.; Gottschalk, S.M.; et al. Safety and clinical efficacy of rapidly-generated trivirus-directed T cells as treatment for adenovirus, EBV, and CMV infections after allogeneic hematopoietic stem cell transplant. Mol. Ther. 2013, 21, 2113–2121. [Google Scholar] [CrossRef] [Green Version]
  17. Blyth, E.; Clancy, L.; Simms, R.; Ma, C.K.K.; Burgess, J.; Deo, S.; Byth, K.; Dubosq, M.-C.; Shaw, P.J.; Micklethwaite, K.P.; et al. Donor-derived CMV-specific T cells reduce the requirement for CMV-directed pharmacotherapy after allogeneic stem cell transplantation. Blood 2013, 121, 3745–3758. [Google Scholar] [CrossRef] [Green Version]
  18. Papadopoulou, A.; Gerdemann, U.; Katari, U.L.; Tzannou, I.; Liu, H.; Martinez, C.; Leung, K.; Carrum, G.; Gee, A.P.; Vera, J.F.; et al. Activity of Broad-Spectrum T Cells as Treatment for AdV, EBV, CMV, BKV, and HHV6 Infections after HSCT. Sci. Transl. Med. 2014, 6, 242ra83. [Google Scholar] [CrossRef] [Green Version]
  19. Heslop, H.E.; Slobod, K.S.; Pule, M.A.; Hale, G.A.; Rousseau, A.; Smith, C.A.; Bollard, C.M.; Liu, H.; Wu, M.-F.; Rochester, R.J.; et al. Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood 2010, 115, 925–935. [Google Scholar] [CrossRef] [Green Version]
  20. Keller, M.D.; Darko, S.; Lang, H.; Ransier, A.; Lazarski, C.A.; Wang, Y.; Hanley, P.J.; Davila, B.J.; Heimall, J.R.; Ambinder, R.F.; et al. T-cell receptor sequencing demonstrates persistence of virus-specific T cells after antiviral immunotherapy. Br. J. Haematol. 2019, 187, 206–218. [Google Scholar] [CrossRef]
  21. Le Bert, N.; Tan, A.T.; Kunasegaran, K.; Tham, C.Y.L.; Hafezi, M.; Chia, A.; Chng, M.H.Y.; Lin, M.; Tan, N.; Linster, M.; et al. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 2020, 584, 457–462. [Google Scholar] [CrossRef] [PubMed]
  22. Pujadas, E.; Chaudhry, F.; McBride, R.; Richter, F.; Zhao, S.; Wajnberg, A.; Nadkarni, G.; Glicksberg, B.S.; Houldsworth, J.; Cordon-Cardo, C. SARS-CoV-2 viral load predicts COVID-19 mortality. Lancet Respir. Med. 2020, 8, e70. [Google Scholar] [CrossRef] [PubMed]
  23. Bergamaschi, L.; Mescia, F.; Turner, L.; Hanson, A.L.; Kotagiri, P.; Dunmore, B.J.; Ruffieux, H.; De Sa, A.; Huhn, O.; Morgan, M.D.; et al. Longitudinal analysis reveals that delayed bystander CD8+ T cell activation and early immune pathology distinguish severe COVID-19 from mild disease. Immunity 2021, 54, 1257–1275.e8. [Google Scholar] [CrossRef] [PubMed]
  24. Hadjadj, J.; Yatim, N.; Barnabei, L.; Corneau, A.; Boussier, J.; Smith, N.; Péré, H.; Charbit, B.; Bondet, V.; Chenevier-Gobeaux, C.; et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 2020, 369, 718–724. [Google Scholar] [CrossRef] [PubMed]
  25. Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.-C.; Uhl, S.; Hoagland, D.; Møller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID19. Cell 2020, 181, 1036–1045.e9. [Google Scholar] [CrossRef] [PubMed]
  26. Moore, B.J.B.; June, C.H. Cytokine release syndrome in severe COVID19. Science 2020, 368, 473–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Humar, A.; St Louis, P.; Mazzulli, T.; McGeer, A.; Lipton, J.; Messner, H.; Macdonald, K.S. Elevated Serum Cytokines Are Associated with Cytomegalovirus Infection and Disease in Bone Marrow Transplant Recipients. J. Infect. Dis. 1999, 179, 484–488. [Google Scholar] [CrossRef] [Green Version]
  28. Ramos-Casals, M.; Brito-Zerón, P.; López-Guillermo, A.; Khamashta, M.A.; Bosch, X. Adult haemophagocytic syndrome. Lancet 2014, 383, 1503–1516. [Google Scholar] [CrossRef]
  29. Basar, R.; Uprety, N.; Ensley, E.; Daher, M.; Klein, K.; Martinez, F.; Aung, F.; Shanley, M.; Hu, B.; Gokdemir, E.; et al. Generation of glucocorticoid-resistant SARS-CoV-2 T cells for adoptive cell therapy. Cell Rep. 2021, 36, 109432. [Google Scholar] [CrossRef]
  30. Takeuchi, O.; Akira, S. Recognition of viruses by innate immunity. Immunol. Rev. 2007, 220, 214–224. [Google Scholar] [CrossRef]
  31. Gibson, P.G.; Qin, L.; Puah, S.H. COVID-19 acute respiratory distress syndrome (ARDS): Clinical features and differences from typical pre-COVID-19 ARDS. Med. J. Aust. 2020, 213, 54–56.e1. [Google Scholar] [CrossRef] [PubMed]
  32. Shah, M.D.; Sumeh, A.S.; Sheraz, M.; Kavitha, M.S.; Maran, B.A.V.; Rodrigues, K.F. A mini-review on the impact of COVID 19 on vital organs. Biomed. Pharmacother. 2021, 143, 112158. [Google Scholar] [CrossRef] [PubMed]
  33. Pierce, C.A.; Preston-Hurlburt, P.; Dai, Y.; Aschner, C.B.; Cheshenko, N.; Galen, B.; Garforth, S.J.; Herrera, N.G.; Jangra, R.K.; Morano, N.C.; et al. Immune responses to SARS-CoV-2 infection in hospitalized pediatric and adult patients. Sci. Transl. Med. 2020, 12, eabd5487. [Google Scholar] [CrossRef] [PubMed]
  34. Cohen, C.A.; Li, A.P.Y.; Hachim, A.; Hui, D.S.C.; Kwan, M.Y.W.; Tsang, O.T.Y.; Chiu, S.S.; Chan, W.H.; Yau, Y.S.; Kavian, N.; et al. SARS-CoV-2 specific T cell responses are lower in children and increase with age and time after infection. Nat. Commun. 2021, 12, 4678. [Google Scholar] [CrossRef] [PubMed]
  35. Chou, J.; Thomas, P.G.; Randolph, A.G. Immunology of SARS-CoV-2 infection in children. Nat. Immunol. 2022, 23, 177–185. [Google Scholar] [CrossRef] [PubMed]
  36. Nikolopoulou, G.B.; Maltezou, H.C. COVID-19 in Children: Where do we Stand? Arch. Med. Res. 2021, 53, 1–8. [Google Scholar] [CrossRef]
  37. Onofrio, L.; Caraglia, M.; Facchini, G.; Margherita, V.; De Placido, S.; Buonerba, C. Toll-like receptors and COVID-19: A two-faced story with an exciting ending. Future Sci. OA 2020, 6, FSO605. [Google Scholar] [CrossRef]
  38. Khanmohammadi, S.; Rezaei, N. Role of Toll-like receptors in the pathogenesis of COVID-19. J. Med. Virol. 2021, 93, 2735–2739. [Google Scholar] [CrossRef]
  39. Choudhury, A.; Mukherjee, S. In silico studies on the comparative characterization of the interactions of SARS-CoV-2 spike glycoprotein with ACE-2 receptor homologs and human TLRs. J. Med. Virol. 2020, 92, 2105–2113. [Google Scholar] [CrossRef]
  40. Bhattacharya, M.; Sharma, A.R.; Mallick, B.; Sharma, G.; Lee, S.-S.; Chakraborty, C. Immunoinformatics approach to understand molecular interaction between multi-epitopic regions of SARS-CoV-2 spike-protein with TLR4/MD-2 complex. Infect. Genet. Evol. 2020, 85, 104587. [Google Scholar] [CrossRef]
  41. Yu, L.; Wang, L.; Chen, S. Endogenous toll-like receptor ligands and their biological significance. J. Cell Mol. Med. 2010, 14, 2592–2603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Fitzgerald, K.; Rowe, D.C.; Barnes, B.J.; Caffrey, D.R.; Visintin, A.; Latz, E.; Monks, B.; Pitha, P.M.; Golenbock, D.T. LPS-TLR4 Signaling to IRF-3/7 and NF-κB Involves the Toll Adapters TRAM and TRIF. J. Exp. Med. 2003, 198, 1043–1055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. O’Neill, L.A.J.; Bowie, A.G. The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat. Rev. Immunol. 2007, 7, 353–364. [Google Scholar] [CrossRef]
  44. Ribero, M.S.; Jouvenet, N.; Dreux, M.; Nisole, S. Interplay between SARS-CoV-2 and the type I interferon response. PLoS Pathog. 2020, 16, e1008737. [Google Scholar] [CrossRef]
  45. Yang, L.; Xie, X.; Tu, Z.; Fu, J.; Xu, D.; Zhou, Y. The signal pathways and treatment of cytokine storm in COVID-19. Signal Transduct. Target. Ther. 2021, 6, 255. [Google Scholar] [CrossRef]
  46. De Wit, E.; Van Doremalen, N.; Falzarano, D.; Munster, V.J. SARS and MERS: Recent insights into emerging coronaviruses. Nat. Rev. Microbiol. 2016, 14, 523–534. [Google Scholar] [CrossRef] [PubMed]
  47. Peiris, J.; Chu, C.; Cheng, V.; Chan, K.; Hung, I.; Poon, L.; Law, K.; Tang, B.; Hon, T.; Chan, C.; et al. Clinical progression and viral load in a community outbreak of coronavirus-associated SARS pneumonia: A prospective study. Lancet 2003, 361, 1767–1772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Wong, R.; Wu, A.; To, K.F.; Lee, N.; Lam, C.W.K.; Wong, C.K.; Chan, P.; Ng, M.H.L.; Yu, L.M.; Hui, D.; et al. Haematological manifestations in patients with severe acute respiratory syndrome: Retrospective analysis. BMJ 2003, 326, 1358–1362. [Google Scholar] [CrossRef] [Green Version]
  49. Galani, I.-E.; Rovina, N.; Lampropoulou, V.; Triantafyllia, V.; Manioudaki, M.; Pavlos, E.; Koukaki, E.; Fragkou, P.C.; Panou, V.; Rapti, V.; et al. Untuned antiviral immunity in COVID-19 revealed by temporal type I/III interferon patterns and flu comparison. Nat. Immunol. 2020, 22, 32–40. [Google Scholar] [CrossRef]
  50. Channappanavar, R.; Perlman, S. Pathogenic human coronavirus infections: Causes and consequences of cytokine storm and immunopathology. Semin. Immunopathol. 2017, 39, 529–539. [Google Scholar] [CrossRef]
  51. Aboudounya, M.M.; Heads, R.J. COVID-19 and Toll-Like Receptor 4 (TLR4): SARS-CoV-2 May Bind and Activate TLR4 to Increase ACE2 Expression, Facilitating Entry and Causing Hyperinflammation. Mediat. Inflamm. 2021, 2021, 8874339. [Google Scholar] [CrossRef] [PubMed]
  52. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.-H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e278. [Google Scholar] [CrossRef] [PubMed]
  53. Ortiz, M.E.; Thurman, A.; Pezzulo, A.A.; Leidinger, M.R.; Klesney-Tait, J.A.; Karp, P.H.; Tan, P.; Wohlford-Lenane, C.; McCray, P.B.; Meyerholz, D.K. Heterogeneous expression of the SARS-Coronavirus-2 receptor ACE2 in the human respiratory tract. eBioMedicine 2020, 60, 102976. [Google Scholar] [CrossRef] [PubMed]
  54. Zou, X.; Chen, K.; Zou, J.; Han, P.; Hao, J.; Han, Z. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front. Med. 2020, 14, 185–192. [Google Scholar] [CrossRef] [Green Version]
  55. Jackson, C.B.; Farzan, M.; Chen, B.; Choe, H. Mechanisms of SARS-CoV-2 entry into cells. Nat. Rev. Mol. Cell Biol. 2021, 23, 3–20. [Google Scholar] [CrossRef]
  56. Ziegler, C.G.K.; Allon, S.J.; Nyquist, S.K.; Mbano, I.M.; Miao, V.N.; Tzouanas, C.N.; Cao, Y.; Yousif, A.S.; Bals, J.; Hauser, B.M.; et al. SARS-CoV-2 Receptor ACE2 Is an Interferon-Stimulated Gene in Human Airway Epithelial Cells and Is Detected in Specific Cell Subsets across Tissues. Cell 2020, 181, 1016–1035.e19. [Google Scholar] [CrossRef]
  57. Zhou, L.; Niu, Z.; Jiang, X.; Zhang, Z.; Zheng, Y.; Wang, Z.; Zhu, Y.; Gao, L.; Huang, H.; Wang, X.; et al. SARS-CoV-2 Targets by the pscRNA Profiling of ACE2, TMPRSS2 and Furin Proteases. iScience 2020, 23, 101744. [Google Scholar] [CrossRef]
  58. Bertram, S.; Heurich, A.; Lavender, H.; Gierer, S.; Danisch, S.; Perin, P.; Lucas, J.M.; Nelson, P.S.; Pöhlmann, S.; Soilleux, E.J. Influenza and SARS-Coronavirus Activating Proteases TMPRSS2 and HAT Are Expressed at Multiple Sites in Human Respiratory and Gastrointestinal Tracts. PLoS ONE 2012, 7, e35876. [Google Scholar] [CrossRef] [Green Version]
  59. Bassi, D.E.; Zhang, J.; Renner, C.; Klein-Szanto, A.J. Targeting proprotein convertases in furin-rich lung cancer cells results in decreased in vitro and in vivo growth. Mol. Carcinog. 2016, 56, 1182–1188. [Google Scholar] [CrossRef]
  60. Coutard, B.; Valle, C.; de Lamballerie, X.; Canard, B.; Seidah, N.G.; Decroly, E. The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antivir. Res. 2020, 176, 104742. [Google Scholar] [CrossRef]
  61. Imai, Y.; Kuba, K.; Rao, S.; Huan, Y.; Guo, F.; Guan, B.; Yang, P.; Sarao, R.; Wada, T.; Leong-Poi, H.; et al. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature 2005, 436, 112–116. [Google Scholar] [CrossRef] [PubMed]
  62. Samavati, L.; Uhal, B.D. ACE2, Much More Than Just a Receptor for SARS-COV-2. Front. Cell Infect. Microbiol. 2020, 10, 317. [Google Scholar] [CrossRef]
  63. Zipeto, D.; Palmeira, J.D.F.; Argañaraz, G.A.; Argañaraz, E.R. ACE2/ADAM17/TMPRSS2 Interplay May Be the Main Risk Factor for COVID-19. Front. Immunol. 2020, 11, 576745. [Google Scholar] [CrossRef]
  64. Gheware, A.; Ray, A.; Rana, D.; Bajpai, P.; Nambirajan, A.; Arulselvi, S.; Mathur, P.; Trikha, A.; Arava, S.; Das, P.; et al. ACE2 protein expression in lung tissues of severe COVID-19 infection. Sci. Rep. 2022, 12, 4058. [Google Scholar] [CrossRef]
  65. Forrester, S.J.; Booz, G.W.; Sigmund, C.D.; Coffman, T.M.; Kawai, T.; Rizzo, V.; Scalia, R.; Eguchi, S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol. Rev. 2018, 98, 1627–1738. [Google Scholar] [CrossRef]
  66. Iwasaki, M.; Saito, J.; Zhao, H.; Sakamoto, A.; Hirota, K.; Ma, D. Inflammation Triggered by SARS-CoV-2 and ACE2 Augment Drives Multiple Organ Failure of Severe COVID-19: Molecular Mechanisms and Implications. Inflammation 2020, 44, 13–34. [Google Scholar] [CrossRef]
  67. Ji, Y.; Liu, J.; Wang, Z.; Liu, N. Angiotensin II Induces Inflammatory Response Partly Via Toll-Like Receptor 4-Dependent Signaling Pathway in Vascular Smooth Muscle Cells. Cell Physiol. Biochem. 2009, 23, 265–276. [Google Scholar] [CrossRef]
  68. Sharif-Askari, N.S.; Sharif-Askari, F.S.; Alabed, M.; Temsah, M.-H.; Al Heialy, S.; Hamid, Q.; Halwani, R. Airways Expression of SARS-CoV-2 Receptor, ACE2, and TMPRSS2 Is Lower in Children Than Adults and Increases with Smoking and COPD. Mol. Ther.—Methods Clin. Dev. 2020, 18, 1–6. [Google Scholar] [CrossRef]
  69. Bunyavanich, S.; Do, A.; Vicencio, A. Nasal Gene Expression of Angiotensin-Converting Enzyme 2 in Children and Adults. JAMA 2020, 323, 2427–2429. [Google Scholar] [CrossRef]
  70. Muus, C.; Luecken, M.; Eraslan, G.; Waghray, A.; Heimberg, G.; Sikkema, L.; Kobayashi, Y.; Vaishnav, E.D.; Subramanian, A.; Smilie, C.; et al. Integrated analyses of single-cell atlases reveal age, gender, and smoking status associations with cell type-specific expression of mediators of SARS-CoV-2 viral entry and highlights inflammatory programs in putative target cells. BioRxiv 2020. [Google Scholar] [CrossRef]
  71. Wang, A.; Chiou, J.; Poirion, O.B.; Buchanan, J.; Valdez, M.J.; Verheyden, J.M.; Hou, X.; Kudtarkar, P.; Narendra, S.; Newsome, J.M.; et al. Single-cell multiomic profiling of human lungs reveals cell-type-specific and age-dynamic control of SARS-CoV2 host genes. eLife 2020, 9, e62522. [Google Scholar] [CrossRef] [PubMed]
  72. Schuler, B.A.; Habermann, A.C.; Plosa, E.J.; Taylor, C.J.; Jetter, C.; Negretti, N.M.; Kapp, M.E.; Benjamin, J.T.; Gulleman, P.; Nichols, D.S.; et al. Age-determined expression of priming protease TMPRSS2 and localization of SARS-CoV-2 in lung epithelium. J. Clin. Investig. 2021, 131, e140766. [Google Scholar] [CrossRef] [PubMed]
  73. Grifoni, A.; Weiskopf, D.; Ramirez, S.I.; Mateus, J.; Dan, J.M.; Moderbacher, C.R.; Rawlings, S.A.; Sutherland, A.; Premkumar, L.; Jadi, R.S.; et al. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020, 181, 1489–1501.e1415. [Google Scholar] [CrossRef] [PubMed]
  74. Luo, L.; Liang, W.; Pang, J.; Xu, G.; Chen, Y.; Guo, X.; Wang, X.; Zhao, Y.; Lai, Y.; Liu, Y.; et al. Dynamics of TCR repertoire and T cell function in COVID-19 convalescent individuals. Cell Discov. 2021, 7, 89. [Google Scholar] [CrossRef] [PubMed]
  75. Shah, K.; Al-Haidari, A.; Sun, J.; Kazi, J.U. T cell receptor (TCR) signaling in health and disease. Signal Transduct. Target. Ther. 2021, 6, 412. [Google Scholar] [CrossRef]
  76. Shatrova, A.N.; Mityushova, E.V.; Vassilieva, I.O.; Aksenov, N.D.; Zenin, V.V.; Nikolsky, N.N.; Marakhova, I.I. Time-Dependent Regulation of IL-2R α-Chain (CD25) Expression by TCR Signal Strength and IL-2-Induced STAT5 Signaling in Activated Human Blood T Lymphocytes. PLoS ONE 2016, 11, e0167215. [Google Scholar] [CrossRef] [Green Version]
  77. Yang, X.; Dai, T.; Zhou, X.; Qian, H.; Guo, R.; Lei, L.; Zhang, X.; Zhang, D.; Shi, L.; Cheng, Y.; et al. Naturally activated adaptive immunity in COVID-19 patients. J. Cell Mol. Med. 2020, 24, 12457–12463. [Google Scholar] [CrossRef]
  78. Sojka, D.K.; Bruniquel, D.; Schwartz, R.H.; Singh, N.J. IL-2 Secretion by CD4+ T Cells In Vivo Is Rapid, Transient, and Influenced by TCR-Specific Competition. J. Immunol. 2004, 172, 6136–6143. [Google Scholar] [CrossRef] [Green Version]
  79. Kalfaoglu, B.; Almeida-Santos, J.; Tye, C.A.; Satou, Y.; Ono, M. T-Cell Hyperactivation and Paralysis in Severe COVID-19 Infection Revealed by Single-Cell Analysis. Front. Immunol. 2020, 11, 589380. [Google Scholar] [CrossRef]
  80. Wu, Y.; Borde, M.; Heissmeyer, V.; Feuerer, M.; Lapan, A.D.; Stroud, J.; Bates, D.L.; Guo, L.; Han, A.; Ziegler, S.F.; et al. FOXP3 Controls Regulatory T Cell Function through Cooperation with NFAT. Cell 2006, 126, 375–387. [Google Scholar] [CrossRef]
  81. Schmidt, A.; Oberle, N.; Krammer, P.H. Molecular Mechanisms of Treg-Mediated T Cell Suppression. Front. Immunol. 2012, 3, 51. [Google Scholar] [CrossRef] [Green Version]
  82. Ono, M. Control of regulatory T-cell differentiation and function by T-cell receptor signalling and Foxp3 transcription factor complexes. Immunology 2020, 160, 24–37. [Google Scholar] [CrossRef] [Green Version]
  83. Hashimoto, M.; Kamphorst, A.O.; Im, S.J.; Kissick, H.T.; Pillai, R.N.; Ramalingam, S.S.; Araki, K.; Ahmed, R. CD8 T Cell Exhaustion in Chronic Infection and Cancer: Opportunities for Interventions. Annu. Rev. Med. 2018, 69, 301–318. [Google Scholar] [CrossRef]
  84. Gao, Y.; Tang, J.; Chen, W.; Li, Q.; Nie, J.; Lin, F.; Wu, Q.; Chen, Z.; Gao, Z.; Fan, H.; et al. Inflammation negatively regulates FOXP3 and regulatory T-cell function via DBC1. Proc. Natl. Acad. Sci. USA 2015, 112, E3246–E3254. [Google Scholar] [CrossRef] [Green Version]
  85. Meckiff, B.J.; Ramírez-Suástegui, C.; Fajardo, V.; Chee, S.J.; Kusnadi, A.; Simon, H.; Eschweiler, S.; Grifoni, A.; Pelosi, E.; Weiskopf, D.; et al. Imbalance of Regulatory and Cytotoxic SARS-CoV-2-Reactive CD4+ T Cells in COVID-19. Cell 2020, 183, 1340–1353.e16. [Google Scholar] [CrossRef]
  86. Ortutay, Z.; Oksanen, A.; Aittomäki, S.; Ortutay, C.; Pesu, M. Proprotein convertase FURIN regulates T cell receptor-induced transactivation. J. Leukoc. Biol. 2015, 98, 73–83. [Google Scholar] [CrossRef]
  87. Rajendiran, A.; Tenbrock, K. Regulatory T cell function in autoimmune disease. J. Transl. Autoimmun. 2021, 4, 100130. [Google Scholar] [CrossRef]
  88. Oft, M. Immune regulation and cytotoxic T cell activation of IL-10 agonists—Preclinical and clinical experience. Semin. Immunol. 2019, 44, 101325. [Google Scholar] [CrossRef]
  89. Smith, L.K.; Boukhaled, G.M.; Condotta, S.A.; Mazouz, S.; Guthmiller, J.J.; Vijay, R.; Butler, N.S.; Bruneau, J.; Shoukry, N.H.; Krawczyk, C.M.; et al. Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity. Immunity 2018, 48, 299–312.e5. [Google Scholar] [CrossRef]
  90. Ejrnaes, M.; Filippi, C.M.; Martinic, M.; Ling, E.M.; Togher, L.M.; Crotty, S.; Von Herrath, M.G. Resolution of a chronic viral infection after interleukin-10 receptor blockade. J. Exp. Med. 2006, 203, 2461–2472. [Google Scholar] [CrossRef]
  91. Blackburn, S.D.; Wherry, E.J. IL-10, T cell exhaustion and viral persistence. Trends Microbiol. 2007, 15, 143–146. [Google Scholar] [CrossRef] [PubMed]
  92. Erickson, J.J.; Lu, P.; Wen, S.; Hastings, A.K.; Gilchuk, P.; Joyce, S.; Shyr, Y.; Williams, J.V. Acute Viral Respiratory Infection Rapidly Induces a CD8+ T Cell Exhaustion–like Phenotype. J. Immunol. 2015, 195, 4319–4330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Lu, L.; Zhang, H.; Dauphars, D.J.; He, Y.-W. A Potential Role of Interleukin 10 in COVID-19 Pathogenesis. Trends Immunol. 2020, 42, 3–5. [Google Scholar] [CrossRef]
  94. De Biasi, S.; Meschiari, M.; Gibellini, L.; Bellinazzi, C.; Borella, R.; Fidanza, L.; Gozzi, L.; Iannone, A.; Tartaro, D.L.; Mattioli, M.; et al. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia. Nat. Commun. 2020, 11, 3434. [Google Scholar] [CrossRef]
  95. Laing, A.G.; Lorenc, A.; del Barrio, I.D.M.; Das, A.; Fish, M.; Monin, L.; Muñoz-Ruiz, M.; McKenzie, D.R.; Hayday, T.S.; Francos-Quijorna, I.; et al. A dynamic COVID-19 immune signature includes associations with poor prognosis. Nat. Med. 2020, 26, 1623–1635. [Google Scholar] [CrossRef] [PubMed]
  96. Schub, D.; Klemis, V.; Schneitler, S.; Mihm, J.; Lepper, P.M.; Wilkens, H.; Bals, R.; Eichler, H.; Gärtner, B.C.; Becker, S.L.; et al. High levels of SARS-CoV-2–specific T cells with restricted functionality in severe courses of COVID-19. J. Clin. Investig. 2020, 5, e142167. [Google Scholar] [CrossRef] [PubMed]
  97. Zhou, Y.; Fu, B.; Zheng, X.; Wang, D.; Zhao, C.; Qi, Y.; Sun, R.; Tian, Z.; Xu, X.; Wei, H. Pathogenic T-cells and inflammatory monocytes incite inflammatory storms in severe COVID-19 patients. Natl. Sci. Rev. 2020, 7, 998–1002. [Google Scholar] [CrossRef] [Green Version]
  98. Schultheiß, C.; Paschold, L.; Simnica, D.; Mohme, M.; Willscher, E.; von Wenserski, L.; Scholz, R.; Wieters, I.; Dahlke, C.; Tolosa, E.; et al. Next-Generation Sequencing of T and B Cell Receptor Repertoires from COVID-19 Patients Showed Signatures Associated with Severity of Disease. Immunity 2020, 53, 442–455.e4. [Google Scholar] [CrossRef]
  99. Zheng, M.; Gao, Y.; Wang, G.; Song, G.; Liu, S.; Sun, D.; Xu, Y.; Tian, Z. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell Mol. Immunol. 2020, 17, 533–535. [Google Scholar] [CrossRef] [Green Version]
  100. Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef]
  101. Angelosanto, J.M.; Blackburn, S.D.; Crawford, A.; Wherry, E.J. Progressive Loss of Memory T Cell Potential and Commitment to Exhaustion during Chronic Viral Infection. J. Virol. 2012, 86, 8161–8170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Brooks, D.G.; McGavern, D.; Oldstone, M.B. Reprogramming of antiviral T cells prevents inactivation and restores T cell activity during persistent viral infection. J. Clin. Investig. 2006, 116, 1675–1685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Utzschneider, D.T.; Charmoy, M.; Chennupati, V.; Pousse, L.; Ferreira, D.P.; Calderon-Copete, S.; Danilo, M.; Alfei, F.; Hofmann, M.; Wieland, D.; et al. T Cell Factor 1-Expressing Memory-like CD8+ T Cells Sustain the Immune Response to Chronic Viral Infections. Immunity 2016, 45, 415–427. [Google Scholar] [CrossRef] [Green Version]
  104. Zheng, H.-Y.; Zhang, M.; Yang, C.-X.; Zhang, N.; Wang, X.-C.; Yang, X.-P.; Dong, X.-Q.; Zheng, Y.-T. Elevated exhaustion levels and reduced functional diversity of T cells in peripheral blood may predict severe progression in COVID-19 patients. Cell Mol. Immunol. 2020, 17, 541–543. [Google Scholar] [CrossRef]
  105. Welch, J.L.; Xiang, J.; Chang, Q.; Houtman, J.C.D.; Stapleton, J.T. T-Cell Expression of Angiotensin-Converting Enzyme 2 and Binding of Severe Acute Respiratory Coronavirus 2. J. Infect. Dis. 2021, 225, 810–819. [Google Scholar] [CrossRef]
  106. Jeannet, R.; Daix, T.; Formento, R.; Feuillard, J.; François, B. Severe COVID-19 is associated with deep and sustained multifaceted cellular immunosuppression. Intensiv. Care Med. 2020, 46, 1769–1771. [Google Scholar] [CrossRef]
  107. Lucas, C.; Wong, P.; Klein, J.; Castro, T.B.R.; Silva, J.; Sundaram, M.; Ellingson, M.K.; Mao, T.; Oh, J.E.; Israelow, B.; et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature 2020, 584, 463–469. [Google Scholar] [CrossRef]
  108. Lu, X.; Zhang, L.; Du, H.; Zhang, J.; Li, Y.Y.; Qu, J.; Zhang, W.; Wang, Y.; Bao, S.; Li, Y.; et al. SARS-CoV-2 Infection in Children. N. Engl. J. Med. 2020, 382, 1663–1665. [Google Scholar] [CrossRef] [Green Version]
  109. Bai, K.; Liu, W.; Liu, C.; Fu, Y.; Hu, J.; Qin, Y.; Zhang, Q.; Chen, H.; Xu, F.; Li, C. Clinical Analysis of 25 COVID-19 Infections in Children. Pediatr. Infect. Dis. J. 2020, 39, e100–e103. [Google Scholar] [CrossRef]
  110. Pierce, C.A.; Sy, S.; Galen, B.; Goldstein, D.Y.; Orner, E.; Keller, M.J.; Herold, K.C.; Herold, B.C. Natural mucosal barriers and COVID-19 in children. JCI Insight 2021, 6, e148694. [Google Scholar] [CrossRef]
  111. Xia, H.; Shi, P.-Y. Antagonism of Type I Interferon by Severe Acute Respiratory Syndrome Coronavirus 2. J. Interf. Cytokine Res. 2020, 40, 543–548. [Google Scholar] [CrossRef] [PubMed]
  112. Bastard, P.; Rosen, L.B.; Zhang, Q.; Michailidis, E.; Hoffmann, H.-H.; Zhang, Y.; Dorgham, K.; Philippot, Q.; Rosain, J.; Béziat, V.; et al. Auto-antibodies against type I IFNs in patients with life-threatening COVID-19. Science 2020, 370, eabd4585. [Google Scholar] [CrossRef]
  113. Chen, J.; Lau, Y.F.; Lamirande, E.W.; Paddock, C.D.; Bartlett, J.H.; Zaki, S.R.; Subbarao, K. Cellular Immune Responses to Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) Infection in Senescent BALB/c Mice: CD4+ T Cells Are Important in Control of SARS-CoV Infection. J. Virol. 2010, 84, 1289–1301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Kong, S.L.; Chui, P.; Lim, B.; Salto-Tellez, M. Elucidating the molecular physiopathology of acute respiratory distress syndrome in severe acute respiratory syndrome patients. Virus Res. 2009, 145, 260–269. [Google Scholar] [CrossRef]
  115. Wong, C.K.; Lam, C.W.K.; Wu, A.K.L.; Ip, W.K.; Lee, N.L.S.; Chan, I.H.S.; Lit, L.C.W.; Hui, D.S.C.; Chan, M.H.M.; Chung, S.S.C.; et al. Plasma inflammatory cytokines and chemokines in severe acute respiratory syndrome. Clin. Exp. Immunol. 2004, 136, 95–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Baas, T.; Taubenberger, J.K.; Chong, P.Y.; Chui, P.; Katze, M.G. SARS-CoV Virus-Host Interactions and Comparative Etiologies of Acute Respiratory Distress Syndrome as Determined by Transcriptional and Cytokine Profiling of Formalin-Fixed Paraffin-Embedded Tissues. J. Interf. Cytokine Res. 2006, 26, 309–317. [Google Scholar] [CrossRef] [Green Version]
  117. Subbarao, K.; Roberts, A. Is there an ideal animal model for SARS? Trends Microbiol. 2006, 14, 299–303. [Google Scholar] [CrossRef]
  118. Channappanavar, R.; Zhao, J.; Perlman, S. T cell-mediated immune response to respiratory coronaviruses. Immunol. Res. 2014, 59, 118–128. [Google Scholar] [CrossRef] [Green Version]
  119. Szabo, P.A.; Dogra, P.; Gray, J.I.; Wells, S.B.; Connors, T.J.; Weisberg, S.P.; Krupska, I.; Matsumoto, R.; Poon, M.M.; Idzikowski, E.; et al. Longitudinal profiling of respiratory and systemic immune responses reveals myeloid cell-driven lung inflammation in severe COVID-19. Immunity 2021, 54, 797–814.e6. [Google Scholar] [CrossRef]
  120. Liao, M.; Liu, Y.; Yuan, J.; Wen, Y.; Xu, G.; Zhao, J.; Cheng, L.; Li, J.; Wang, X.; Wang, F.; et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 2020, 26, 842–844. [Google Scholar] [CrossRef]
  121. Bange, E.M.; Han, N.A.; Wileyto, P.; Kim, J.Y.; Gouma, S.; Robinson, J.; Greenplate, A.R.; Hwee, M.A.; Porterfield, F.; Owoyemi, O.; et al. CD8+ T cells contribute to survival in patients with COVID-19 and hematologic cancer. Nat. Med. 2021, 27, 1280–1289. [Google Scholar] [CrossRef] [PubMed]
  122. Moderbacher, C.R.; Ramirez, S.I.; Dan, J.M.; Grifoni, A.; Hastie, K.M.; Weiskopf, D.; Belanger, S.; Abbott, R.K.; Kim, C.; Choi, J.; et al. Antigen-Specific Adaptive Immunity to SARS-CoV-2 in Acute COVID-19 and Associations with Age and Disease Severity. Cell 2020, 183, 996–1012.e19. [Google Scholar] [CrossRef] [PubMed]
  123. Dan, J.M.; Mateus, J.; Kato, Y.; Hastie, K.M.; Yu, E.D.; Faliti, C.E.; Grifoni, A.; Ramirez, S.I.; Haupt, S.; Frazier, A.; et al. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science 2021, 371, eabf4063. [Google Scholar] [CrossRef]
  124. Le Bert, N.; Clapham, H.E.; Tan, A.T.; Ni Chia, W.; Tham, C.Y.; Lim, J.M.; Kunasegaran, K.; Tan, L.W.L.; Dutertre, C.-A.; Shankar, N.; et al. Highly functional virus-specific cellular immune response in asymptomatic SARS-CoV-2 infection. J. Exp. Med. 2021, 218, e20202617. [Google Scholar] [CrossRef]
  125. Tan, A.T.; Linster, M.; Tan, C.W.; Le Bert, N.; Ni Chia, W.; Kunasegaran, K.; Zhuang, Y.; Tham, C.Y.L.; Chia, A.; Smith, G.J.D.; et al. Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients. Cell Rep. 2021, 34, 108728. [Google Scholar] [CrossRef] [PubMed]
  126. Agha, M.E.; Blake, M.; Chilleo, C.; Wells, A.; Haidar, G. Suboptimal Response to Coronavirus Disease 2019 Messenger RNA Vaccines in Patients With Hematologic Malignancies: A Need for Vigilance in the Postmasking Era. Open Forum Infect. Dis. 2021, 8, ofab353. [Google Scholar] [CrossRef] [PubMed]
  127. Barrière, J.; Chamorey, E.; Adjtoutah, Z.; Castelnau, O.; Mahamat, A.; Marco, S.; Petit, E.; Leysalle, A.; Raimondi, V.; Carles, M. Impaired immunogenicity of BNT162b2 anti-SARS-CoV-2 vaccine in patients treated for solid tumors. Ann. Oncol. 2021, 32, 1053–1055. [Google Scholar] [CrossRef]
  128. Hagin, D.; Freund, T.; Navon, M.; Halperin, T.; Adir, D.; Marom, R.; Levi, I.; Benor, S.; Alcalay, Y.; Freund, N.T. Immunogenicity of Pfizer-BioNTech COVID-19 vaccine in patients with inborn errors of immunity. J. Allergy Clin. Immunol. 2021, 148, 739–749. [Google Scholar] [CrossRef]
  129. Mamez, A.-C.; Pradier, A.; Giannotti, F.; Petitpas, A.; Urdiola, M.F.; Vu, D.-L.; Masouridi-Levrat, S.; Morin, S.; Dantin, C.; Clerc-Renaud, D.; et al. Antibody responses to SARS-CoV2 vaccination in allogeneic hematopoietic stem cell transplant recipients. Bone Marrow Transplant. 2021, 56, 3094–3096. [Google Scholar] [CrossRef]
  130. Delmonte, O.M.; Bergerson, J.R.; Burbelo, P.D.; Durkee-Shock, J.R.; Dobbs, K.; Bosticardo, M.; Keller, M.D.; McDermott, D.H.; Rao, V.K.; Dimitrova, D.; et al. Antibody responses to the SARS-CoV-2 vaccine in individuals with various inborn errors of immunity. J. Allergy Clin. Immunol. 2021, 148, 1192–1197. [Google Scholar] [CrossRef]
  131. Apostolidis, S.A.; Kakara, M.; Painter, M.M.; Goel, R.R.; Mathew, D.; Lenzi, K.; Rezk, A.; Patterson, K.R.; Espinoza, D.A.; Kadri, J.C.; et al. Cellular and humoral immune responses following SARS-CoV-2 mRNA vaccination in patients with multiple sclerosis on anti-CD20 therapy. Nat. Med. 2021, 27, 1990–2001. [Google Scholar] [CrossRef] [PubMed]
  132. Naik, S.; Nicholas, S.K.; Martinez, C.A.; Leen, A.M.; Hanley, P.J.; Gottschalk, S.M.; Rooney, C.M.; Hanson, I.C.; Krance, R.A.; Shpall, E.J.; et al. Adoptive immunotherapy for primary immunodeficiency disorders with virus-specific T lymphocytes. J. Allergy Clin. Immunol. 2016, 137, 1498–1505.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Ng, O.-W.; Chia, A.; Tan, A.T.; Jadi, R.S.; Leong, H.N.; Bertoletti, A.; Tan, Y.-J. Memory T cell responses targeting the SARS coronavirus persist up to 11 years post-infection. Vaccine 2016, 34, 2008–2014. [Google Scholar] [CrossRef] [PubMed]
  134. Zhao, J.; Alshukairi, A.N.; Baharoon, S.A.; Ahmed, W.A.; Bokhari, A.A.; Nehdi, A.M.; Layqah, L.A.; Alghamdi, M.G.; Al Gethamy, M.M.; Dada, A.M.; et al. Recovery from the Middle East respiratory syndrome is associated with antibody and T cell responses. Sci. Immunol. 2017, 2, eaan5393. [Google Scholar] [CrossRef] [Green Version]
  135. Jung, J.H.; Rha, M.-S.; Sa, M.; Choi, H.K.; Jeon, J.H.; Seok, H.; Park, D.W.; Park, S.-H.; Jeong, H.W.; Choi, W.S.; et al. SARS-CoV-2-specific T cell memory is sustained in COVID-19 convalescent patients for 10 months with successful development of stem cell-like memory T cells. Nat. Commun. 2021, 12, 4043. [Google Scholar] [CrossRef]
  136. Noh, J.Y.; Yang, J.-S.; Hwang, S.Y.; Hyun, H.; Seong, H.; Yoon, J.G.; Yoon, S.-Y.; Cheong, H.J.; Kim, W.J.; Park, W.-J.; et al. Duration of Humoral Immunity and Cross-Neutralizing Activity Against the Alpha, Beta, and Delta Variants After Wild-Type Severe Acute Respiratory Syndrome Coronavirus 2 Infection: A Prospective Cohort Study. J. Infect. Dis. 2022, 226, 975–978. [Google Scholar] [CrossRef]
  137. Sukdolak, C.; Tischer, S.; Dieks, D.; Figueiredo, C.; Goudeva, L.; Heuft, H.-G.; Verboom, M.; Immenschuh, S.; Heim, A.; Borchers, S.; et al. CMV-, EBV- and ADV-Specific T Cell Immunity: Screening and Monitoring of Potential Third-Party Donors to Improve Post-Transplantation Outcome. Biol. Blood Marrow Transplant. 2013, 19, 1480–1492. [Google Scholar] [CrossRef] [Green Version]
  138. Tischer, S.; Priesner, C.; Heuft, H.-G.; Goudeva, L.; Mende, W.; Barthold, M.; Kloeß, S.; Arseniev, L.; Aleksandrova, K.; Maecker-Kolhoff, B.; et al. Rapid generation of clinical-grade antiviral T cells: Selection of suitable T-cell donors and GMP-compliant manufacturing of antiviral T cells. J. Transl. Med. 2014, 12, 336. [Google Scholar] [CrossRef] [Green Version]
  139. Conway, S.R.; Keller, M.D.; Bollard, C.M. Cellular therapies for the treatment and prevention of SARS-CoV-2 infection. Blood 2022, 140, 208–221. [Google Scholar] [CrossRef]
  140. Tischer, S.; Kaireit, T.; Figueiredo, C.; Hiller, O.; Maecker-Kolhoff, B.; Geyeregger, R.; Immenschuh, S.; Blasczyk, R.; Eiz-Vesper, B. Establishment of the reversible peptide-major histocompatibility complex (pMHC) class I Histamer technology: Tool for visualization and selection of functionally active antigen-specific CD8+ T lymphocytes. Int. Immunol. 2012, 24, 561–572. [Google Scholar] [CrossRef]
  141. Rauser, G.; Einsele, H.; Sinzger, C.; Wernet, D.; Kuntz, G.; Assenmacher, M.; Campbell, J.D.M.; Topp, M.S. Rapid generation of combined CMV-specific CD4+ and CD8+ T-cell lines for adoptive transfer into recipients of allogeneic stem cell transplants. Blood 2004, 103, 3565–3572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Cobbold, M.; Khan, N.; Pourgheysari, B.; Tauro, S.; McDonald, D.; Osman, H.; Assenmacher, M.; Billingham, L.; Steward, C.; Crawley, C.; et al. Adoptive transfer of cytomegalovirus-specific CTL to stem cell transplant patients after selection by HLA–peptide tetramers. J. Exp. Med. 2005, 202, 379–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Committee for Advanced Therapies of the European Medicines Agency. Available online: www.ema.europa.eu/docs/en_GB/document_library/Report/2013/05/WC500143582.pdf (accessed on 9 September 2022).
  144. Von Rossum, A.; Krall, R.; Escalante, N.; Choy, J.C. Inflammatory Cytokines Determine the Susceptibility of Human CD8 T Cells to Fas-mediated Activation-induced Cell Death through Modulation of FasL and c-FLIPS Expression. J. Biol. Chem. 2011, 286, 21137–21144. [Google Scholar] [CrossRef] [Green Version]
  145. Leung, W.; Soh, T.G.; Linn, Y.C.; Low, J.G.; Loh, J.; Chan, M.; Chng, W.J.; Koh, L.P.; Poon, M.L.; Ng, K.P.; et al. Rapid production of clinical-grade SARS-CoV-2 specific T cells. Adv. Cell Gene Ther. 2020, 3, e101. [Google Scholar] [CrossRef] [PubMed]
  146. Cooper, R.S.; Fraser, A.R.; Smith, L.; Burgoyne, P.; Imlach, S.N.; Jarvis, L.M.; Turner, D.M.; Zahra, S.; Turner, M.L.; Campbell, J.D.M. Rapid GMP-Compliant Expansion of SARS-CoV-2–Specific T Cells From Convalescent Donors for Use as an Allogeneic Cell Therapy for COVID-19. Front. Immunol. 2021, 11, 598402. [Google Scholar] [CrossRef] [PubMed]
  147. Guerreiro, M.; Aguilar-Gallardo, C.; Montoro, J.; Francés-Gómez, C.; Latorre, V.; Luna, I.; Planelles, D.; Carrasco, M.P.; Gómez, M.D.; González-Barberá, E.M.; et al. Adoptive transfer of ex vivo expanded SARS-CoV-2-specific cytotoxic lymphocytes: A viable strategy for COVID-19 immunosuppressed patients? Transpl. Infect. Dis. 2021, 23, e13602. [Google Scholar] [CrossRef]
  148. García-Ríos, E.; Leivas, A.; Mancebo, F.J.; Sánchez-Vega, L.; Lanzarot, D.; Aguado, J.M.; Martínez-López, J.; Paciello, M.L.; Pérez-Romero, P. Isolation of Functional SARS-CoV-2 Antigen-Specific T-Cells with Specific Viral Cytotoxic Activity for Adoptive Therapy of COVID-19. Biomedicines 2022, 10, 630. [Google Scholar] [CrossRef]
  149. Bonifacius, A.; Tischer-Zimmermann, S.; Santamorena, M.M.; Mausberg, P.; Schenk, J.; Koch, S.; Barnstorf-Brandes, J.; Gödecke, N.; Martens, J.; Goudeva, L.; et al. Rapid Manufacturing of Highly Cytotoxic Clinical-Grade SARS-CoV-2-specific T Cell Products Covering SARS-CoV-2 and Its Variants for Adoptive T Cell Therapy. Front. Bioeng. Biotechnol. 2022, 10, 867042. [Google Scholar] [CrossRef]
  150. Franzke, A.; Piao, W.; Lauber, J.; Gatzlaff, P.; Könecke, C.; Hansen, W.; Schmitt-Thomsen, A.; Hertenstein, B.; Buer, J.; Ganser, A. G-CSF as immune regulator in T cells expressing the G-CSF receptor: Implications for transplantation and autoimmune diseases. Blood 2003, 102, 734–739. [Google Scholar] [CrossRef] [Green Version]
  151. Toh, H.C.; Sun, L.; Soe, Y.; Wu, Y.; Phoon, Y.P.; Chia, W.K.; Wu, J.; Wong, K.Y.; Tan, P. G-CSF induces a potentially tolerant gene and immunophenotype profile in T cells in vivo. Clin. Immunol. 2009, 132, 83–92. [Google Scholar] [CrossRef] [PubMed]
  152. Bunse, C.E.; Borchers, S.; Varanasi, P.R.; Tischer, S.; Figueiredo, C.; Immenschuh, S.; Kalinke, U.; Koehl, U.; Goudeva, L.; Maecker-Kolhoff, B.; et al. Impaired Functionality of Antiviral T Cells in G-CSF Mobilized Stem Cell Donors: Implications for the Selection of CTL Donor. PLoS ONE 2013, 8, e77925. [Google Scholar] [CrossRef]
  153. Uhlin, M.; Gertow, J.; Uzunel, M.; Okas, M.; Berglund, S.; Watz, E.; Brune, M.; Ljungman, P.; Maeurer, M.; Mattsson, J. Rapid Salvage Treatment With Virus-Specific T Cells for Therapy-Resistant Disease. Clin. Infect. Dis. 2012, 55, 1064–1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Leen, A.M.; Bollard, C.M.; Mendizabal, A.M.; Shpall, E.J.; Szabolcs, P.; Antin, J.H.; Kapoor, N.; Pai, S.-Y.; Rowley, S.D.; Kebriaei, P.; et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood 2013, 121, 5113–5123. [Google Scholar] [CrossRef] [PubMed]
  155. Gallot, G.; Vollant, S.; Saïagh, S.; Clémenceau, B.; Vivien, R.; Cerato, E.; Bignon, J.-D.; Ferrand, C.; Jaccard, A.; Vigouroux, S.; et al. T-cell Therapy Using a Bank of EBV-specific Cytotoxic T Cells: Lessons from a phase I/II feasibility and safety study. J. Immunother. 2014, 37, 170–179. [Google Scholar] [CrossRef]
  156. Withers, B.; Blyth, E.; Clancy, L.E.; Yong, A.; Fraser, C.; Burgess, J.; Simms, R.; Brown, R.; Kliman, D.; Dubosq, M.-C.; et al. Long-term control of recurrent or refractory viral infections after allogeneic HSCT with third-party virus-specific T cells. Blood Adv. 2017, 1, 2193–2205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Melenhorst, J.J.; Leen, A.M.; Bollard, C.M.; Quigley, M.F.; Price, D.A.; Rooney, C.M.; Brenner, M.K.; Barrett, A.J.; Heslop, H.E. Allogeneic virus-specific T cells with HLA alloreactivity do not produce GVHD in human subjects. Blood 2010, 116, 4700–4702. [Google Scholar] [CrossRef]
  158. Doubrovina, E.; Oflaz-Sozmen, B.; Prockop, S.E.; Kernan, N.A.; Abramson, S.; Teruya-Feldstein, J.; Hedvat, C.; Chou, J.F.; Heller, G.; Barker, J.N.; et al. Adoptive immunotherapy with unselected or EBV-specific T cells for biopsy-proven EBV+ lymphomas after allogeneic hematopoietic cell transplantation. Blood 2012, 119, 2644–2656. [Google Scholar] [CrossRef]
  159. Icheva, V.; Kayser, S.; Wolff, D.; Tuve, S.; Kyzirakos, C.; Bethge, W.; Greil, J.; Albert, M.H.; Schwinger, W.; Nathrath, M.; et al. Adoptive Transfer of Epstein-Barr Virus (EBV) Nuclear Antigen 1–Specific T Cells As Treatment for EBV Reactivation and Lymphoproliferative Disorders After Allogeneic Stem-Cell Transplantation. J. Clin. Oncol. 2013, 31, 39–48. [Google Scholar] [CrossRef]
  160. O’Reilly, R.J.; Prockop, S.; Hasan, A.N.; Koehne, G.; Doubrovina, E. Virus-specific T-cell banks for ‘off the shelf’ adoptive therapy of refractory infections. Bone Marrow Transplant. 2016, 51, 1163–1172. [Google Scholar] [CrossRef] [Green Version]
  161. Tzannou, I.; Watanabe, A.; Naik, S.; Daum, R.; Kuvalekar, M.; Leung, K.S.; Martinez, C.; Sasa, G.; Wu, M.; Gee, A.P.; et al. “Mini” bank of only 8 donors supplies CMV-directed T cells to diverse recipients. Blood Adv. 2019, 3, 2571–2580. [Google Scholar] [CrossRef]
  162. Vasileiou, S.; Kuvalekar, M.; Workineh, A.; Watanabe, A.; Velazquez, Y.; Lulla, S.; Heslop, H.E.; Mooney, K.; Grimes, K.; Carrum, G.; et al. 37. Allogeneic, Off-the-Shelf, SARS-CoV-2-specific T Cells Demonstrate Reactivity Against Emerging Variant Strains. Open Forum Infect. Dis. 2021, 8, S27. [Google Scholar] [CrossRef]
  163. Braun, J.; Loyal, L.; Frentsch, M.; Wendisch, D.; Georg, P.; Kurth, F.; Hippenstiel, S.; Dingeldey, M.; Kruse, B.; Fauchere, F.; et al. SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19. Nature 2020, 587, 270–274. [Google Scholar] [CrossRef]
  164. Keller, M.D.; Harris, K.M.; Jensen-Wachspress, M.A.; Kankate, V.V.; Lang, H.; Lazarski, C.A.; Durkee-Shock, J.; Lee, P.-H.; Chaudhry, K.; Webber, K.; et al. SARS-CoV-2–specific T cells are rapidly expanded for therapeutic use and target conserved regions of the membrane protein. Blood 2020, 136, 2905–2917. [Google Scholar] [CrossRef]
  165. Mateus, J.; Grifoni, A.; Tarke, A.; Sidney, J.; Ramirez, S.I.; Dan, J.M.; Burger, Z.C.; Rawlings, S.A.; Smith, D.M.; Phillips, E.; et al. Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science 2020, 370, 89–94. [Google Scholar] [CrossRef]
  166. Goel, R.R.; Painter, M.M.; Apostolidis, S.A.; Mathew, D.; Meng, W.; Rosenfeld, A.M.; Lundgreen, K.A.; Reynaldi, A.; Khoury, D.S.; Pattekar, A.; et al. mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science 2021, 374, abm0829. [Google Scholar] [CrossRef]
  167. Tarke, A.; Sidney, J.; Methot, N.; Yu, E.D.; Zhang, Y.; Dan, J.M.; Goodwin, B.; Rubiro, P.; Sutherland, A.; Wang, E.; et al. Impact of SARS-CoV-2 variants on the total CD4+ and CD8+ T cell reactivity in infected or vaccinated individuals. Cell Rep. Med. 2021, 2, 100355. [Google Scholar] [CrossRef]
  168. Painter, M.M.; Mathew, D.; Goel, R.R.; Apostolidis, S.A.; Pattekar, A.; Kuthuru, O.; Baxter, A.E.; Herati, R.S.; Oldridge, D.A.; Gouma, S.; et al. Rapid induction of antigen-specific CD4+ T cells is associated with coordinated humoral and cellular immunity to SARS-CoV-2 mRNA vaccination. Immunity 2021, 54, 2133–2142.e3. [Google Scholar] [CrossRef]
  169. Stanojevic, M.; Geiger, A.; Ostermeier, B.; Sohai, D.; Lazarski, C.; Lang, H.; Jensen-Wachspress, M.; Webber, K.; Burbelo, P.; Cohen, J.; et al. Spike-directed vaccination elicits robust spike-specific T-cell response, including to mutant strains. Cytotherapy 2021, 24, 10–15. [Google Scholar] [CrossRef]
  170. Mazzoni, A.; Vanni, A.; Spinicci, M.; Capone, M.; Lamacchia, G.; Salvati, L.; Coppi, M.; Antonelli, A.; Carnasciali, A.; Farahvachi, P.; et al. SARS-CoV-2 Spike-Specific CD4+ T Cell Response Is Conserved Against Variants of Concern, Including Omicron. Front. Immunol. 2022, 13, 801431. [Google Scholar] [CrossRef]
  171. Taborska, P.; Lastovicka, J.; Stakheev, D.; Strizova, Z.; Bartunkova, J.; Smrz, D. SARS-CoV-2 spike glycoprotein-reactive T cells can be readily expanded from COVID-19 vaccinated donors. Immun. Inflamm. Dis. 2021, 9, 1452–1467. [Google Scholar] [CrossRef]
  172. Grifoni, A.; Sidney, J.; Vita, R.; Peters, B.; Crotty, S.; Weiskopf, D.; Sette, A. SARS-CoV-2 human T cell epitopes: Adaptive immune response against COVID-19. Cell Host Microbe 2021, 29, 1076–1092. [Google Scholar] [CrossRef]
  173. Tarke, A.; Sidney, J.; Kidd, C.K.; Dan, J.M.; Ramirez, S.I.; Yu, E.D.; Mateus, J.; da Silva Antunes, R.; Moore, E.; Rubiro, P.; et al. Comprehensive analysis of T cell immunodominance and immunoprevalence of SARS-CoV-2 epitopes in COVID-19 cases. Cell Rep. Med. 2021, 2, 100204. [Google Scholar] [CrossRef]
  174. Moss, P. The T cell immune response against SARS-CoV-2. Nat. Immunol. 2022, 23, 186–193. [Google Scholar] [CrossRef]
  175. Quadeer, A.A.; Ahmed, S.F.; McKay, M.R. Landscape of epitopes targeted by T cells in 852 individuals recovered from COVID-19: Meta-analysis, immunoprevalence, and web platform. Cell Rep. Med. 2021, 2, 100312. [Google Scholar] [CrossRef]
  176. Hamelin, D.J.; Fournelle, D.; Grenier, J.-C.; Schockaert, J.; Kovalchik, K.A.; Kubiniok, P.; Mostefai, F.; Duquette, J.D.; Saab, F.; Sirois, I.; et al. The mutational landscape of SARS-CoV-2 variants diversifies T cell targets in an HLA-supertype-dependent manner. Cell Syst. 2021, 13, 143–157.e3. [Google Scholar] [CrossRef]
  177. Skelly, D.T.; Harding, A.C.; Gilbert-Jaramillo, J.; Knight, M.L.; Longet, S.; Brown, A.; Adele, S.; Adland, E.; Brown, H.; Tipton, T.; et al. Two doses of SARS-CoV-2 vaccination induce robust immune responses to emerging SARS-CoV-2 variants of concern. Nat. Commun. 2021, 12, 5061. [Google Scholar] [CrossRef]
  178. Kuhlmann, C.; Mayer, C.K.; Claassen, M.; Maponga, T.; Burgers, W.A.; Keeton, R.; Riou, C.; Sutherland, A.D.; Suliman, T.; Shaw, M.L.; et al. Breakthrough infections with SARS-CoV-2 omicron despite mRNA vaccine booster dose. Lancet 2022, 399, 625–626. [Google Scholar] [CrossRef]
  179. GeurtsvanKessel, C.H.; Geers, D.; Schmitz, K.S.; Mykytyn, A.Z.; Lamers, M.M.; Bogers, S.; Scherbeijn, S.; Gommers, L.; Sablerolles, R.S.G.; Nieuwkoop, N.N.; et al. Divergent SARS-CoV-2 Omicron–reactive T and B cell responses in COVID-19 vaccine recipients. Sci. Immunol. 2022, 7, eabo2202. [Google Scholar] [CrossRef]
  180. Zhang, H.; Deng, S.; Ren, L.; Zheng, P.; Hu, X.; Jin, T.; Tan, X. Profiling CD8+ T cell epitopes of COVID-19 convalescents reveals reduced cellular immune responses to SARS-CoV-2 variants. Cell Rep. 2021, 36, 109708. [Google Scholar] [CrossRef]
  181. de Silva, T.I.; Liu, G.; Lindsey, B.B.; Dong, D.; Moore, S.C.; Hsu, N.S.; Shah, D.; Wellington, D.; Mentzer, A.J.; Angyal, A.; et al. The impact of viral mutations on recognition by SARS-CoV-2 specific T cells. iScience 2021, 24, 103353. [Google Scholar] [CrossRef]
  182. The WHO Rapid Evidence Appraisal for COVID-19 Therapies (REACT) Working Group; Sterne, J.A.C.; Murthy, S.; Diaz, J.V.; Slutsky, A.S.; Villar, J.; Angus, D.C.; Annane, D.; Azevedo, L.C.P.; Berwanger, O.; et al. Association Between Administration of Systemic Corticosteroids and Mortality Among Critically Ill Patients With COVID-19: A Meta-analysis. JAMA 2020, 324, 1330–1341. [Google Scholar] [CrossRef]
  183. Li, H.; Yan, B.; Gao, R.; Ren, J.; Yang, J. Effectiveness of corticosteroids to treat severe COVID-19: A systematic review and meta-analysis of prospective studies. Int. Immunopharmacol. 2021, 100, 108121. [Google Scholar] [CrossRef]
  184. Braat, M.C.; Oosterhuis, B.; Koopmans, R.P.; Meewis, J.M.; Van Boxtel, C.J. Kinetic-dynamic modeling of lymphocytopenia induced by the combined action of dexamethasone and hydrocortisone in humans, after inhalation and intravenous administration of dexamethasone. J. Pharmacol. Exp. Ther. 1992, 262, 509–515. [Google Scholar]
  185. Ma, J.; Xie, Y.; Shi, Y.; Qin, W.; Zhao, B.; Jin, Y. Glucocorticoid-induced apoptosis requires FOXO3A activity. Biochem. Biophys. Res. Commun. 2008, 377, 894–898. [Google Scholar] [CrossRef]
  186. Orange, J.S. Natural killer cell deficiency. J. Allergy Clin. Immunol. 2013, 132, 515–525. [Google Scholar] [CrossRef] [Green Version]
  187. Lisco, A.; Hsu, A.P.; Dimitrova, D.; Proctor, D.M.; Mace, E.M.; Ye, P.; Anderson, M.V.; Hicks, S.N.; Grivas, C.; Hammoud, D.A.; et al. Treatment of Relapsing HPV Diseases by Restored Function of Natural Killer Cells. New Engl. J. Med. 2021, 385, 921–929. [Google Scholar] [CrossRef]
  188. Wilk, A.J.; Rustagi, A.; Zhao, N.Q.; Roque, J.; Martínez-Colón, G.J.; McKechnie, J.L.; Ivison, G.T.; Ranganath, T.; Vergara, R.; Hollis, T.; et al. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 2020, 26, 1070–1076. [Google Scholar] [CrossRef]
  189. Wang, F.; Nie, J.; Wang, H.; Zhao, Q.; Xiong, Y.; Deng, L.; Song, S.; Ma, Z.; Mo, P.; Zhang, Y. Characteristics of Peripheral Lymphocyte Subset Alteration in COVID-19 Pneumonia. J. Infect. Dis. 2020, 221, 1762–1769. [Google Scholar] [CrossRef] [Green Version]
  190. Jiang, Y.; Wei, X.; Guan, J.; Qin, S.; Wang, Z.; Lu, H.; Qian, J.; Wu, L.; Chen, Y.; Chen, Y.; et al. COVID-19 pneumonia: CD8+ T and NK cells are decreased in number but compensatory increased in cytotoxic potential. Clin. Immunol. 2020, 218, 108516. [Google Scholar] [CrossRef]
  191. Osman, M.; Faridi, R.M.; Sligl, W.; Shabani-Rad, M.-T.; Dharmani-Khan, P.; Parker, A.; Kalra, A.; Tripathi, M.B.; Storek, J.; Tervaert, J.W.C.; et al. Impaired natural killer cell counts and cytolytic activity in patients with severe COVID-19. Blood Adv. 2020, 4, 5035–5039. [Google Scholar] [CrossRef]
  192. Littwitz, E.; Francois, S.; Dittmer, U.; Gibbert, K. Distinct roles of NK cells in viral immunity during different phases of acute Friend retrovirus infection. Retrovirology 2013, 10, 127. [Google Scholar] [CrossRef]
  193. Ahmed, F.; Jo, D.-H.; Lee, S.-H. Can Natural Killer Cells Be a Principal Player in Anti-SARS-CoV-2 Immunity? Front. Immunol. 2020, 11, 586765. [Google Scholar] [CrossRef]
  194. Van Eeden, C.; Khan, L.; Osman, M.S.; Tervaert, J.W.C. Natural Killer Cell Dysfunction and its Role in COVID-19. Int. J. Mol. Sci. 2020, 21, 6351. [Google Scholar] [CrossRef]
  195. Björkström, N.K.; Strunz, B.; Ljunggren, H.-G. Natural killer cells in antiviral immunity. Nat. Rev. Immunol. 2021, 22, 112–123. [Google Scholar] [CrossRef]
  196. Zavvar, M.; Yahyapoor, A.; Baghdadi, H.; Zargaran, S.; Assadiasl, S.; Abdolmohammadi, K.; Abooei, A.H.; Sattarian, M.R.; JalaliFarahani, M.; Zarei, N.; et al. COVID-19 immunotherapy: Treatment based on the immune cell-mediated approaches. Int. Immunopharmacol. 2022, 107, 108655. [Google Scholar] [CrossRef]
  197. Ma, M.T.; Badeti, S.; Chen, C.-H.; Kim, J.; Choudhary, A.; Honnen, B.; Reichman, C.; Calianese, D.; Pinter, A.; Jiang, Q.; et al. CAR-NK Cells Effectively Target SARS-CoV-2-Spike-Expressing Cell Lines In Vitro. Front. Immunol. 2021, 12, 652223. [Google Scholar] [CrossRef]
  198. D’Alessio, F.R.; Tsushima, K.; Aggarwal, N.R.; West, E.E.; Willett, M.H.; Britos, M.F.; Pipeling, M.R.; Brower, R.G.; Tuder, R.M.; McDyer, J.F.; et al. CD4+CD25+Foxp3+ Tregs resolve experimental lung injury in mice and are present in humans with acute lung injury. J. Clin. Investig. 2009, 119, 2898–2913. [Google Scholar] [CrossRef] [Green Version]
  199. Fulton, R.B.; Meyerholz, D.K.; Varga, S.M. Foxp3+ CD4 Regulatory T Cells Limit Pulmonary Immunopathology by Modulating the CD8 T Cell Response during Respiratory Syncytial Virus Infection. J. Immunol. 2010, 185, 2382–2392. [Google Scholar] [CrossRef] [Green Version]
  200. Arpaia, N.; Green, J.A.; Moltedo, B.; Arvey, A.; Hemmers, S.; Yuan, S.; Treuting, P.M.; Rudensky, A.Y. A Distinct Function of Regulatory T Cells in Tissue Protection. Cell 2015, 162, 1078–1089. [Google Scholar] [CrossRef] [Green Version]
  201. Wilson, J.G.; Liu, K.D.; Zhuo, H.; Caballero, L.; McMillan, M.; Fang, X.; Cosgrove, K.; Vojnik, R.; Calfee, C.S.; Lee, J.-W.; et al. Mesenchymal stem (stromal) cells for treatment of ARDS: A phase 1 clinical trial. Lancet Respir. Med. 2014, 3, 24–32. [Google Scholar] [CrossRef] [Green Version]
  202. Hu, S.; Li, J.; Xu, X.; Liu, A.; He, H.; Xu, J.; Chen, Q.; Liu, S.; Liu, L.; Qiu, H.; et al. The hepatocyte growth factor-expressing character is required for mesenchymal stem cells to protect the lung injured by lipopolysaccharide in vivo. Stem Cell Res. Ther. 2016, 7, 66. [Google Scholar] [CrossRef] [PubMed]
  203. Matthay, M.A.; Calfee, C.S.; Zhuo, H.; Thompson, B.T.; Wilson, J.G.; Levitt, J.E.; Rogers, A.J.; Gotts, J.E.; Wiener-Kronish, J.P.; Bajwa, E.K.; et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): A randomised phase 2a safety trial. Lancet Respir. Med. 2018, 7, 154–162. [Google Scholar] [CrossRef] [PubMed]
  204. Leng, Z.; Zhu, R.; Hou, W.; Feng, Y.; Yang, Y.; Han, Q.; Shan, G.; Meng, F.; Du, D.; Wang, S.; et al. Transplantation of ACE2- Mesenchymal Stem Cells Improves the Outcome of Patients with COVID-19 Pneumonia. Aging Dis. 2020, 11, 216–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Avanzini, M.A.; Mura, M.; Percivalle, E.; Bastaroli, F.; Croce, S.; Valsecchi, C.; Lenta, E.; Nykjaer, G.; Cassaniti, I.; Bagnarino, J.; et al. Human mesenchymal stromal cells do not express ACE2 and TMPRSS2 and are not permissive to SARS-CoV-2 infection. Stem Cells Transl. Med. 2020, 10, 636–642. [Google Scholar] [CrossRef] [PubMed]
  206. Hernandez, J.J.; Beaty, D.E.; Fruhwirth, L.L.; Chaves, A.P.L.; Riordan, N.H. Dodging COVID-19 infection: Low expression and localization of ACE2 and TMPRSS2 in multiple donor-derived lines of human umbilical cord-derived mesenchymal stem cells. J. Transl. Med. 2021, 19, 149. [Google Scholar] [CrossRef] [PubMed]
  207. Vasanthan, J.; Gurusamy, N.; Rajasingh, S.; Sigamani, V.; Kirankumar, S.; Thomas, E.L.; Rajasingh, J. Role of Human Mesenchymal Stem Cells in Regenerative Therapy. Cells 2020, 10, 54. [Google Scholar] [CrossRef] [PubMed]
  208. Le Burel, S.; Thepenier, C.; Boutin, L.; Lataillade, J.-J.; Peltzer, J. Effect of Mesenchymal Stromal Cells on T Cells in a Septic Context: Immunosuppression or Immunostimulation? Stem Cells Dev. 2017, 26, 1477–1489. [Google Scholar] [CrossRef]
  209. Shi, Y.; Wang, Y.; Li, Q.; Liu, K.; Hou, J.; Shao, C.; Wang, Y. Immunoregulatory mechanisms of mesenchymal stem and stromal cells in inflammatory diseases. Nat. Rev. Nephrol. 2018, 14, 493–507. [Google Scholar] [CrossRef]
  210. Mishra, D.K.; Rocha, H.J.; Miller, R.; Kim, M.P. Immune cells inhibit the tumor metastasis in the 4D cellular lung model by reducing the number of live circulating tumor cells. Sci. Rep. 2018, 8, 16569. [Google Scholar] [CrossRef] [Green Version]
  211. Jeyaraman, M.; John, A.; Koshy, S.; Ranjan, R.; Anudeep, T.C.; Jain, R.; Swati, K.; Jha, N.K.; Sharma, A.; Kesari, K.K.; et al. Fostering mesenchymal stem cell therapy to halt cytokine storm in COVID-19. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2020, 1867, 166014. [Google Scholar] [CrossRef]
  212. Wang, L.; Li, Y.; Xu, M.; Deng, Z.; Zhao, Y.; Yang, M.; Liu, Y.; Yuan, R.; Sun, Y.; Zhang, H.; et al. Regulation of Inflammatory Cytokine Storms by Mesenchymal Stem Cells. Front. Immunol. 2021, 12, 726909. [Google Scholar] [CrossRef] [PubMed]
  213. Doorn, J.; Moll, G.; Le Blanc, K.; van Blitterswijk, C.; de Boer, J. Therapeutic Applications of Mesenchymal Stromal Cells: Paracrine Effects and Potential Improvements. Tissue Eng. Part B Rev. 2012, 18, 101–115. [Google Scholar] [CrossRef] [PubMed]
  214. Bernardo, M.E.; Fibbe, W.E. Mesenchymal Stromal Cells: Sensors and Switchers of Inflammation. Cell Stem Cell 2013, 13, 392–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Shen, Q.; Chen, B.; Xiao, Z.; Zhao, L.; Xu, X.; Wan, X.; Jin, M.; Dai, J.; Dai, H. Paracrine factors from mesenchymal stem cells attenuate epithelial injury and lung fibrosis. Mol. Med. Rep. 2014, 11, 2831–2837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Klyushnenkova, E.; Mosca, J.D.; Zernetkina, V.; Majumdar, M.K.; Beggs, K.J.; Simonetti, D.W.; Deans, R.J.; McIntosh, K.R. T cell responses to allogeneic human mesenchymal stem cells: Immunogenicity, tolerance, and suppression. J. Biomed. Sci. 2005, 12, 47–57. [Google Scholar] [CrossRef]
  217. Grau-Vorster, M.; Laitinen, A.; Nystedt, J.; Vives, J. HLA-DR expression in clinical-grade bone marrow-derived multipotent mesenchymal stromal cells: A two-site study. Stem Cell Res. Ther. 2019, 10, 164. [Google Scholar] [CrossRef] [Green Version]
  218. Wang, Y.; Huang, J.; Gong, L.; Yu, D.; An, C.; Bunpetch, V.; Dai, J.; Huang, H.; Zou, X.; Ouyang, H.; et al. The Plasticity of Mesenchymal Stem Cells in Regulating Surface HLA-I. iScience 2019, 15, 66–78. [Google Scholar] [CrossRef] [Green Version]
  219. Shu, L.; Niu, C.; Li, R.; Huang, T.; Wang, Y.; Huang, M.; Ji, N.; Zheng, Y.; Chen, X.; Shi, L.; et al. Treatment of severe COVID-19 with human umbilical cord mesenchymal stem cells. Stem Cell Res. Ther. 2020, 11, 361. [Google Scholar] [CrossRef]
  220. Guo, Z.; Chen, Y.; Luo, X.; He, X.; Zhang, Y.; Wang, J. Administration of umbilical cord mesenchymal stem cells in patients with severe COVID-19 pneumonia. Crit. Care 2020, 24, 420. [Google Scholar] [CrossRef]
  221. Chen, X.; Shan, Y.; Wen, Y.; Sun, J.; Du, H. Mesenchymal stem cell therapy in severe COVID-19: A retrospective study of short-term treatment efficacy and side effects. J. Infect. 2020, 81, 647–679. [Google Scholar] [CrossRef]
  222. Meng, F.; Xu, R.; Wang, S.; Xu, Z.; Zhang, C.; Li, Y.; Yang, T.; Shi, L.; Fu, J.; Jiang, T.; et al. Human umbilical cord-derived mesenchymal stem cell therapy in patients with COVID-19: A phase 1 clinical trial. Signal Transduct. Target. Ther. 2020, 5, 172. [Google Scholar] [CrossRef] [PubMed]
  223. Lanzoni, G.; Linetsky, E.; Correa, D.; Cayetano, S.M.; Alvarez, R.A.; Kouroupis, D.; Gil, A.A.; Poggioli, R.; Ruiz, P.; Marttos, A.C.; et al. Umbilical cord mesenchymal stem cells for COVID-19 acute respiratory distress syndrome: A double-blind, phase 1/2a, randomized controlled trial. Stem Cells Transl. Med. 2021, 10, 660–673. [Google Scholar] [CrossRef] [PubMed]
  224. Dilogo, I.H.; Aditianingsih, D.; Sugiarto, A.; Burhan, E.; Damayanti, T.; Sitompul, P.A.; Mariana, N.; Antarianto, R.D.; Liem, I.K.; Kispa, T.; et al. Umbilical Cord Mesenchymal Stromal Cells as Critical COVID-19 Adjuvant Therapy: A Randomized Controlled Trial. Stem Cells Transl. Med. 2021, 10, 1279–1287. [Google Scholar] [CrossRef] [PubMed]
  225. Shi, L.; Huang, H.; Lu, X.; Yan, X.; Jiang, X.; Xu, R.; Wang, S.; Zhang, C.; Yuan, X.; Xu, Z.; et al. Effect of human umbilical cord-derived mesenchymal stem cells on lung damage in severe COVID-19 patients: A randomized, double-blind, placebo-controlled phase 2 trial. Signal Transduct. Target. Ther. 2021, 6, 58. [Google Scholar] [CrossRef] [PubMed]
  226. Eiro, N.; Cabrera, J.R.; Fraile, M.; Costa, L.; Vizoso, F.J. The Coronavirus Pandemic (SARS-CoV-2): New Problems Demand New Solutions, the Alternative of Mesenchymal (Stem) Stromal Cells. Front. Cell Dev. Biol. 2020, 8, 645. [Google Scholar] [CrossRef]
  227. Sengupta, V.; Sengupta, S.; Lazo, A.; Woods, P.; Nolan, A.; Bremer, N. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells as Treatment for Severe COVID-19. Stem Cells Dev. 2020, 29, 747–754. [Google Scholar] [CrossRef]
  228. Wing, J.B.; Tanaka, A.; Sakaguchi, S. Human FOXP3+ Regulatory T Cell Heterogeneity and Function in Autoimmunity and Cancer. Immunity 2019, 50, 302–316. [Google Scholar] [CrossRef] [Green Version]
  229. Romano, M.; Fanelli, G.; Albany, C.J.; Giganti, G.; Lombardi, G. Past, Present, and Future of Regulatory T Cell Therapy in Transplantation and Autoimmunity. Front. Immunol. 2019, 10, 43. [Google Scholar] [CrossRef] [Green Version]
  230. Sledzinska, A.; Mucha, M.V.D.; Sledzi, A.; Bergerhoff, K.; Jenner, R.G.; Peggs, K.S.; Quezada, S.A.; Mucha, M.V.D.; Bergerhoff, K.; Hotblack, A.; et al. Regulatory T Cells Restrain Interleukin-2- and Blimp- 1-Dependent Acquisition of Cytotoxic Function by Article Regulatory T Cells Restrain Interleukin-2- and Blimp-1-Dependent Acquisition of Cytotoxic Function by CD4 + T Cells. Immunity 2020, 52, 151–166.e6. [Google Scholar] [CrossRef]
  231. Qin, C.; Zhou, L.; Hu, Z.; Zhang, S.; Yang, S.; Tao, Y.; Xie, C.; Ma, K.; Shang, K.; Wang, W.; et al. Dysregulation of Immune Response in Patients With Coronavirus 2019 (COVID-19) in Wuhan, China. Clin. Infect. Dis. 2020, 71, 762–768. [Google Scholar] [CrossRef]
  232. Chen, G.; Wu, D.; Guo, W.; Cao, Y.; Huang, D.; Wang, H.; Wang, T.; Zhang, X.; Chen, H.; Yu, H.; et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J. Clin. Investig. 2020, 130, 2620–2629. [Google Scholar] [CrossRef]
  233. Wang, F.; Hou, H.; Luo, Y.; Tang, G.; Wu, S.; Huang, M.; Liu, W.; Zhu, Y.; Lin, Q.; Mao, L.; et al. The laboratory tests and host immunity of COVID-19 patients with different severity of illness. J. Clin. Investig. 2020, 5, e137799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Wang, W.; Su, B.; Pang, L.; Qiao, L.; Feng, Y.; Ouyang, Y.; Guo, X.; Shi, H.; Wei, F.; Su, X.; et al. High-dimensional immune profiling by mass cytometry revealed immunosuppression and dysfunction of immunity in COVID-19 patients. Cell Mol. Immunol. 2020, 17, 650–652. [Google Scholar] [CrossRef] [PubMed]
  235. Rahimzadeh, M.; Naderi, N. Toward an understanding of regulatory T cells in COVID-19: A systematic review. J. Med. Virol. 2021, 93, 4167–4181. [Google Scholar] [CrossRef] [PubMed]
  236. Anghelina, D.; Zhao, J.; Trandem, K.; Perlman, S. Role of regulatory T cells in coronavirus-induced acute encephalitis. Virology 2009, 385, 358–367. [Google Scholar] [CrossRef] [Green Version]
  237. Gladstone, D.E.; Kim, B.S.; Mooney, R.K.; Karaba, A.H.; D’Alessio, F.R. Regulatory T Cells for Treating Patients With COVID-19 and Acute Respiratory Distress Syndrome: Two Case Reports. Ann. Intern. Med. 2020, 173, 852–853. [Google Scholar] [CrossRef]
Figure 1. Mechanism of cytokine storm development in COVID-19. ACE2—angiotensin-converting enzyme 2, ADAM17—A disintegrin and metalloproteinase 17, dsRNA—double-stranded RNA, IFNβ—interferon beta, IRF3—interferon regulatory factor 3, MAL—MyD88 adaptor-like protein, MyD88—myeloid differentiation primary response protein 88, NF-κB—nuclear factor kappa B, ssRNA—single-stranded ribonucleic acid, TIR—Toll/interleukin receptor, TLR—Toll-like receptor, TMPRSS2—transmembrane serine protease 2, TRAM—TRIF-related adaptor molecule, TRIF—TIR domain-containing adaptor-inducing interferon-β.
Figure 1. Mechanism of cytokine storm development in COVID-19. ACE2—angiotensin-converting enzyme 2, ADAM17—A disintegrin and metalloproteinase 17, dsRNA—double-stranded RNA, IFNβ—interferon beta, IRF3—interferon regulatory factor 3, MAL—MyD88 adaptor-like protein, MyD88—myeloid differentiation primary response protein 88, NF-κB—nuclear factor kappa B, ssRNA—single-stranded ribonucleic acid, TIR—Toll/interleukin receptor, TLR—Toll-like receptor, TMPRSS2—transmembrane serine protease 2, TRAM—TRIF-related adaptor molecule, TRIF—TIR domain-containing adaptor-inducing interferon-β.
Ijms 23 15122 g001
Figure 2. ACE/Ang2/AT1R and ACE2/Ang-(1-7)/MasR axis during COVID-19. ACE—angiotensin-converting enzyme, ACE2—angiotensin-converting enzyme 2, ADAM17—A disintegrin and metalloproteinase 17, Ang—angiotensin, AT1R—angiotensin II receptor type 1, MasR—Mas receptor, NF-κB—nuclear factor kappa B.
Figure 2. ACE/Ang2/AT1R and ACE2/Ang-(1-7)/MasR axis during COVID-19. ACE—angiotensin-converting enzyme, ACE2—angiotensin-converting enzyme 2, ADAM17—A disintegrin and metalloproteinase 17, Ang—angiotensin, AT1R—angiotensin II receptor type 1, MasR—Mas receptor, NF-κB—nuclear factor kappa B.
Ijms 23 15122 g002
Figure 3. Dysregulation of T cells in COVID-19. APC—antigen-presenting cell, CD25—cluster of differentiation 25, FOXP3—forkhead box-P3, IL—interleukin, IL-2R—interleukin 2 receptor, IL-10R—interleukin 10 receptor.
Figure 3. Dysregulation of T cells in COVID-19. APC—antigen-presenting cell, CD25—cluster of differentiation 25, FOXP3—forkhead box-P3, IL—interleukin, IL-2R—interleukin 2 receptor, IL-10R—interleukin 10 receptor.
Ijms 23 15122 g003
Table 1. SARS-CoV-2-specific T-cell clinical trials for treatment or prevention of severe COVID-19.
Table 1. SARS-CoV-2-specific T-cell clinical trials for treatment or prevention of severe COVID-19.
Clinical TrialTherapyPopulation EligibilityStudy DesignPhase of Clinical TrialEstimated AdmissionBlood Disorder Exclusion
NCT04457726Allogeneic CSTs1 to 90 y
SARS-CoV-2 RT-PCR 1 within 72 h of enrollment
Arm A: severe COVID-19
Arm B: mild to moderate COVID-19 with high risk of progression to severe disease based on age and/
or underlying comorbidity
2/2 (recruiting)18Not excluded unless receiving 0.5 mg/kg/d steroids
NCT04765449Partially HLA-matched
banked allogeneic
CSTs
≥18 y
SARS-CoV-2 RT-PCR 1
High risk of severe disease based on age and/or underlying comorbidity
No supplemental O2 requirement
Arm A: Treatment with CSTs
Arm B: No available HLA-matched
product, monitored at home and
may receive any standard of care
treatment for COVID-19
1 (recruiting)24Included: chemotherapy for
malignancy within the prior 24 mo
Excluded: prior allogeneic HSCT or
solid organ transplant; current
chemotherapy, radiation, and/or
immunosuppressive drug regimen
NCT04742595Partially HLA-matched
banked allogeneic
CSTs
≥18 y
Immunocompromised with cancer
SARS-CoV-2 RT-PCR 1 within 2 wk
of enrollment
Presence of respiratory symptoms
Patients receive CSTs on day 1 and
treatment may repeat every 14 d
at investigators’ discretion.
Early 1 (recruiting)16No (required for inclusion)
NCT04762186Allogeneic CSTsMaximum 14 d between symptom
onset and enrollment
WHO score 5 or 4 with one
additional risk factor for progression
Phase I: dose finding phase
Phase II: randomized pilot study
comparing CST treatment at dose
determined in phase I to current
institutional standard of care
treatment for COVID-19
1/2 (recruiting)12No (inclusion criteria as risk factor for severe disease)
NCT04896606Family derived HLAmatched
Allogeneic CSTs
18 to 65 y
SARS-CoV-2 RT-PCR 1
Hospitalized for mild to moderate COVID-19 disease
Risk of progression based on underlying comorbidity
HLA-matched family related donor with recent SARS-CoV-2 infection at least 10 d from symptom onset available
Experimental arm: Patients receive family donor derived CSTs up to five times every 2 wk along with standard of care.
Active Comparator: Standard of care alone.
2/2 (recruiting)50No (inclusion criteria as risk factor for severe disease)
NCT05141058HSCT donor-derived
allogeneic CSTs
≥12 y and <80 y
≥28 d and <4 wk after allogeneic HSCT
SARS-CoV-2 RT-PCR negative
Arm A: Adults (≥18 to <80 y) will
receive a single infusion of CSTs
at escalating doses for prophylaxis
against SARS-CoV-2 infection.
Arm B: Children (≥12 and <18 y) will receive a single infusion of CSTs at escalating doses for prophylaxis against SARS-CoV-2.
2/2 (recruiting)24No (required for inclusion)
NCT04351659Blood donation from convalescent donor21 to 65 y
history of COVID-19 with documented positive test for SARS-CoV-2 in the past; recovered from COVID-19 and is now suitable for blood donation, fulfilling all standard blood donor criteria; Negative test for SARS-CoV-2 currently
Patients receive blood donation from convalescent donor (1 unit)1 (recruiting)8Yes (Do not meet the standard criteria for blood cell donation)
NCT05447013CoV-2-STs generated from COVID-19recovered donors18 to 80 yHospitalized patients, SARS-CoV-2 PCR positive, within 8 days from the onset of the symptoms who have:
Pneumonia or/and SatO2 ≤ 94% on room air or/and respiratory rate ≥ 24 breaths/min AND
Lymphopenia CD3+ ≤ 650/μL or/and ALC ≤ 1000/microl AND
Increased values of D-dimers (≥2Χ) or/and ferritin (>1000 ng/mL) or/and CRP (≥3Χ) or/and LDH (≥2Χ)
Experimental: For Phase II: Arm A
Standard of care (SOC) and Coronavirus-specific T cells (CoV-2-STs)
Active Comparator:
For Phase II: Arm B
Standard of care (SOC)
2/2 (recruiting)182No
Source: https://clinicaltrials.gov/ct2/home (accessed on 15 August 2022).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nova, Z.; Zemanek, T.; Botek, N. Antigen-Specific T Cells and SARS-CoV-2 Infection: Current Approaches and Future Possibilities. Int. J. Mol. Sci. 2022, 23, 15122. https://doi.org/10.3390/ijms232315122

AMA Style

Nova Z, Zemanek T, Botek N. Antigen-Specific T Cells and SARS-CoV-2 Infection: Current Approaches and Future Possibilities. International Journal of Molecular Sciences. 2022; 23(23):15122. https://doi.org/10.3390/ijms232315122

Chicago/Turabian Style

Nova, Zuzana, Tomas Zemanek, and Norbert Botek. 2022. "Antigen-Specific T Cells and SARS-CoV-2 Infection: Current Approaches and Future Possibilities" International Journal of Molecular Sciences 23, no. 23: 15122. https://doi.org/10.3390/ijms232315122

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

Article Metrics

Back to TopTop