Peribunyaviridae
The
Peribunyaviridae family currently encompasses seven genera and 219 virus species. Among these, the
Orthobunyavirus genus, including the California (CSG), Simbu (SSG), and Bunyamwera serogroups, are the most extensively studied for their ability to cause human infection [
26,
27]. This review specifically focuses on the adaptive immune response to orthobunyaviruses.
The characterization of immunodominant T cell epitopes is lacking for most orthobunyaviruses. Previous studies have primarily observed T cell responses against specific protein subunits, mainly GPC and/or N [
28,
29]. Immunoinformatic studies have identified and predicted virus-specific CD4+ and CD8+ T cell epitopes within GPC and N for Jamestown Canyon (JCV), Oropouche Virus (OROV), and Bunyamwera virus (BUNV), showing high affinity with human major histocompatibility complex class I (MHC-I) and MHC-II (
Table 1) [
28,
29,
30,
31]. While these studies suggest the potential development and use of multi-epitope vaccines, future in vivo studies are required to validate immunogenicity, efficacy, and protection. Notably, the immunogenicity of glycoproteins and N has been confirmed in mice lacking interferon alpha/beta receptors (IFNAR
−/−) for La Crosse virus (LACV) and Schmallenberg virus (SBV) (
Table 1) [
32,
33]. Ex vivo studies confirming T cell responses to entire proteomes has not been conducted for any virus in this family.
In the case of LACV, DNA vaccination in IFNAR
−/− mice with LACV Gn/Gc induced complete protection mediated by CD4+ T cells, while N DNA vaccination provided partial protection [
33]. Similar findings were observed for SBV vaccination, where DNA encoding the Gc ectodomain induced CD8+ T cell-mediated protection [
32]. Additionally, subunit or DNA-based N vaccines have both been shown to reduce clinical signs and significantly decrease viremia upon SBV challenge, eliciting CD8+ T cell-mediated responses [
32,
34].
While most studies on T cell responses to orthobunyaviruses have been conducted using animal models, human T cell responses remain understudied. Notably, LACV’s clinical important has led to comprehensive studies on cellular responses, especially considering its status as the leading cause of pediatric viral encephalitis in the United States [
62]. Children exhibit higher susceptibility to LACV infection, while adults typically experience asymptomatic or mild infections. This age-related susceptibility can be recapitulated in murine models, providing opportunities to study innate and adaptive immune responses against LACV and other related orthobunyaviruses.
In wild-type C57BL/6 mice, both CD4+ and CD8+ T cells (and B cells) play a role in LACV infection [
63]. Although these cell types did not impact neurological disease in weanling mice (3-4 weeks old), they were crucial for protecting adult mice (6-8 weeks old) from LACV pathogenesis [
63]. Natural killer (NK) cells did not appear to have a major role in protection against LACV as their depletion in adult mice did not impact pathogenesis [
63]. These findings underscore the importance of the adaptive immune response in preventing LACV neurological disease, beyond the innate immune response alone.
Given the limited information on T cell responses and immunodominant epitopes, progress in vaccine development for viruses within the Peribunyaviridae family is hampered. Characterization of adaptive immune responses to natural infection, along with in vivo validation of computationally predicted vaccine peptides, is essential to overcome this gap.
Phenuiviridae
The
Phenuiviridae family currently encompasses twenty-two genera and 151 virus species, demonstrating a broad host range that includes humans, animals, plants, and fungi [
64]. In 2018, the World Health Organization identified Rift Valley fever virus (RVFV) and severe fever with thrombocytopenia syndrome virus (SFTSV) as emerging threats, underscoring the urgency for accelerated research and development efforts [
65]. RVFV, transmitted by mosquitoes, is prevalent across Africa and the Middle East. Although typically inducing mild, self-limiting disease, severe complications such as hepatitis, encephalitis, or death can occur [
66]. SFTSV, transmitted by ticks, causes a highly fatal condition marked by hemorrhagic symptoms [
67]. Toscana virus (TOSV), another re-emerging member of this family, ranks among the top etiological agents of aseptic meningitis, and is transmitted by sand flies [
68].
Efforts have been made to identify RVFV, SFTSV, and TOSV epitopes targeted by T cells. Using immunoinformatic approaches, TOSV studies identified T cells epitopes within Gn, Gn, and N, leaving the rest of the viral proteome with unknown immunogenicity [
35]. In contrast the entire proteomes of RVFV and SFTSV was analyzed for immunogenicity, revealing immunodominant T cell epitopes within glycoproteins and N, but also nonstructural proteins (
Table 1) [
36,
37].
In humans vaccinated with formalin-inactivated RVFV, CD4+ and CD8+ T cell targets within Gn, Gc, and N were confirmed in ex vivo assays (
Table 1) [
38]. Another research group defined two HLA-A-2-directed RVFV N epitopes using N-transduced dendritic cells (DCs) to prime CD8+ T cells from HLA-A2 donors [
39]. In vivo assays of T cells responses in mice vaccinated with attenuated RVFV strains also demonstrated that two epitopes within N were robustly targeted (
Table 1) [
40]. For SFTSV, immunoinformatic screening suggested peptides within RdRp and glycoproteins, leading to the in silico evaluation of a multi-epitope vaccine (
Table 1) [
36]. The same approach predicted CD4+ T cell responses against epitopes within the TOSV N and glycoproteins as being highly immunogenic (
Table 1) [
35]. However, in vivo evaluation of epitope-specific T cells from humans vaccinated or infected with SFTSV or TOSV has not been performed.
The immunogenicity of Gn, Gc, and N for RVFV, SFTSV and TOSV has been confirmed with vaccine studies in animal models, emphasizing a protective role for virus-specific CD4+ and CD8+ T cells [
69,
70,
71,
72,
73,
74,
75,
76,
77]. Gn immunization in goats using a recombinant Capripoxvirus vaccine induced protection against RVFV challenge, mediated through a CD4+ T cell response [
69]. BALB/c mice vaccinated with a single dose of a DNA vaccine encoding the RVFV Gn/Gc showed no viraemia or clinical disease, with glycoprotein-specific CD8+ T cell responses, while N-based vaccination conferred only partial protection [
70]. Notably, this vaccine regimen failed to protect IFNAR
-/- mice from RVFV lethal infection, suggesting the involvement of innate immunity in protection [
70]. For SFTSV, vaccine studies involving ferrets and IFNAR
-/- mice revealed that a DNA plasmid encoding Gn/Gc induced protection primarily through antigen-specific T cell responses [
72,
73,
74,
75,
76]. This effect occurred in the absence of detectable antibodies against surface glycoproteins in immunized mice [
73]. The same antigens, when produced via mRNA vaccination, induced a balanced Th1/Th2 response in mice [
74,
75]. Similarly, BALB/c mice were fully protected from a lethal dose of TOSV when vaccinated with recombinant Gc and N (but not when vaccinated with single antigens), demonstrating a potent CD8+ T cell response associated with significant IFN-γ expression [
77].
Furthermore, studies in C57BL/6 mice showed that CD4+ T cells, largely Type 1 T helper cells (Th1)/T follicular helper cells (Tfh) subtypes, play a protective role, with T-box transcription factor TBX21 (T-bet), Cluster of Differentiation 40 (CD40), Cluster of Differentiation 40 Ligand (CD40L), and MCH II pathways crucial in mediating defense against RVFV encephalitis [
40]. In a separate study using immunocompetent mice, infection with an attenuated RVFV strain induced expansion of NK cells, monocytes, and both CD4+ and CD8+ T cells [
78]. Depleting C57Bl/6 mice of CD4+ and CD8+ T cells increased the frequency of encephalitis, supporting that these cell types contribute to the prevention of disease [
78,
79]. It is worth noting that adaptive immune responses against RVFV, due to its rapid progression and high lethality in rodent models, have mostly been explored using attenuated strains or recombinant viral proteins [
40,
78,
80,
81,
82]. In contrast, T cell responses to SFTSV have been well studied in human patients. Non-surviving patients exhibit decreased cells counts, including CD3+, CD4+, and CD8+ T cells, suggesting immune dysfunction in SFTSV disease progression [
83,
84]. CD4+ T cell deficiency and Th1/Th2 imbalance correlate with increased viral load, serum enzymes, cytokines, and disease severity [
85,
86,
87,
88]. Surviving patients have increased expression of activation markers in T cells [
89]. Specifically, CD8+ T cells exhibit a proliferative activated phenotype demonstrated by increased expression of CD69 and CD25, secreting higher level of IFN-γ and granzyme B with enhanced antiviral responses, further supporting that cellular responses play a protective role against infection [
90].
Studies on human T cell responses against TOSV are lacking, as well as to other clinically important viruses within the Phenuiviridae family, including Heartland virus (HRTV), Arumowot virus (AMTV), Uukuniemi virus (UUKV), Guertu virus (GTV), Punta Toro virus (PTV), and sandfly fever Sicilian virus (SFSV). Future studies are needed to better understand cellular immunity against these globally relevant pathogens, aiming to identify correlates of protective immunity that will aid the development of vaccines.
Hantaviridae
The
Hantaviridae family encompasses seven genera and 54 species, responsible for diverse human diseases. Old World hantaviruses in Asia and Europe cause hemorrhagic fever with renal syndrome (HFRS), while New World hantaviruses in North and South America induce hantavirus cardiopulmonary syndrome (HCPS) [
91]. The role of the adaptive immune response in either protection or pathogenesis remains a topic of ongoing investigation [
92,
93].
Immunoinformatic studies have aimed to identify immunodominant T cell epitopes within hantavirus proteins, with the majority revealing epitopes in N followed by the glycoproteins (
Table 1) [
41,
47,
94]. When T cell responses against entire proteomes were analyzed for orthohantaviruses, epitopes within glycoproteins, N, and RdRp and other non-structural proteins were predicted to have high immunogenicity (
Table 1) [
41]. In 1999, the first demonstration of human T cell responses to Hantan virus (HTNV) suggested that CD8+ T cells elicited upon infection are limited to N, recognizing two immunodominant epitopes [
42]. Advances in the field in later years revealed an expanded panel of immunodominant epitopes within the HTNV N in HFRS patients (
Table 1) [
43,
44,
95]. Further detailed characterization suggested HTNV N epitopes restricted by various human leukocyte antigens (HLAs), conserved in both HTNV and Sin Nombre virus (SNV) (
Table 1) [
45,
46]. Moreover, cross-reactivity of N-specific CD8+ T cells against several hantaviruses has been reported in human studies [
42,
44,
96]. Additionally, multiple observations of Gn- and Gc-specific T cell responses have been reported in patients infected with HTNV and Andes virus (ANDV) (
Table 1) [
47,
48]. ANDV epitopes located within the Gn carboxyl-terminus were immunodominant as compared to those from within N and Gc in HCPS patients, and CD8+ T cells targeting ANDV Gn acquire a long-lasting effector phenotype [
48]. CD8+ T cells from patients infected with Puumala virus (PUUV) also exhibit strong responses against a recombinant vaccinia virus expressing N and the second half of Gn [
49]. Notably, virus-specific CD8+ T cell responses during HFRS play a crucial role in HTNV clearance, being efficient releasers of cytotoxic mediators, adopting a memory effector phenotype and their recruitment at early stage of HFRS [
95,
97,
98,
99]. Similarly, an increase of both CD4+ and CD8+ T cells across disease stages correlate with delayed viral clearance in HCPS patients [
100,
101].
The involvement of CD8+ T cells in hantavirus infection is not fully elucidated. Findings in human patients demonstrated a proportional increase in circulating HTNV-infected CD8+ T cells and disease severity [
102]. A recent study in HCPS patients observed an increase in both CD4+ and CD8+ T cells across disease stages, correlating with delayed viral clearance, while in HFRS, the frequency of HTNV-specific effector CD8+ T cells is higher during mild stages compared to the acute phase [
100,
101]. While CD4+ T cell responses have received less attention, studies suggest a mixed Th1/Th2 profile based on cytokine profiles in HTNV infected human sera [
103,
104]. However, there is no clear correlation between effector CD4+ T cells and clinical outcomes.
Insights into protective hantavirus-specific T cell responses have also been obtained from antigen immunization using animal models. BALB/c mice vaccinated with
E. coli-expressed PUUV N developed proliferative Th cells that secrete immune modulators [
105]. The HTNV N and glycoprotein derived immunodominant epitopes previously identified using in silico methods were used to immunize HLA-A2.1/K(b) transgenic mice, both inducing protective T cell responses [
47,
106,
107] Notably, immunization with a multi-epitope HTNV vaccine containing subunits of both N and glycoprotein produced stronger T cell responses compared to single immunization with either epitope in both human cells and transgenic mice [
108,
109].
N protein is relatively conserved and highly immunogenic among hantaviruses [
105,
110,
111,
112]. Given this observation, a study demonstrated cross-protective immune responses against PUUV, Topografov virus (TOPV), ANDV, and Dobrava virus (DOBV) by immunizing bank voles with recombinant N (rN) from different hantaviruses [
113]. When rN immunized mice were challenged against PUUV, cellular responses were more instrumental than humoral response in this cross-protective immunity [
113]. Based on this cross-reactivity study and all the previously mentioned findings, a universal T cell-based vaccine targeting multiple viruses might be achievable and promising in the case of hantaviruses. Additionally, given that multiple immunodominant epitopes within N, Gn, and Gc have been identified in different studies, an unbiased screening of T cell responses against conserved regions of the hantavirus proteome may enable the narrowing down of immunodominant targets that could be useful for cross-protective vaccine development.
Nairoviridae
The
Nairoviridae family currently encompasses three genera and 58 virus species. These viruses are maintained in arthropods and transmitted primarily by ticks to mammals, birds, and bats. Among them, the most significant human pathogen is Crimean-Congo hemorrhagic fever virus (CCHFV), prevalent in Asia, Africa, and Southern and Eastern Europe [
114]. Nairobi sheep disease virus (NSDV) is also noteworthy within this family due to its veterinary impact, causing highly lethal disease in small ruminants in Africa and India [
115].
Research efforts to better understand adaptive immune responses against CCHFV have addressed notable gaps [
116]. Using immunoinformatic approaches, several studies identified CD4+ and CD8+ T cell specific epitopes within CCHFV GPC, N, and RdRp proteins (
Table 1) [
50,
51,
117]. In silico analysis further pinpointed six regions of the CCHFV glycoprotein with high antigenic potential [
50]. The epitope "DCSSTPPDR" in the RdRp was also identified as particularly immunogenic (
Table 1) [
52]. Furthermore, CCHFV survivors demonstrated strong IFN-γ responses against the NSm region of the GP38 protein in ex vivo assays (
Table 1) [
53]. Another study of CCHFV survivors identified cellular responses against N, indicating a preference for non-Gn/Gc epitopes [
54]. Confirming these human findings, immunodominant epitopes were also identified in the N-terminus of Gc followed by NSm as the primary CD8+ T cell targets in CCHFV-infected mice [
55].
Vaccine studies have also supported a role for protective T cell responses against CCHFV challenge [
118,
119]. Mice vaccinated with DNA encoding the CCHFV GPC protected against disease, mediated primarily by CD8+ T cells [
118]. However, in a separate study, signal transducer and activator of transcription 1 knockout (STAT1
-/-) mice immunized with the Gn and Gc ectodomains failed to protect against disease upon CCHFV challenge, even with detectable serum neutralizing antibodies (nAbs) [
120]. In a separate study, IFNAR
-/- mice immunized with nucleoside-modified mRNA-lipid nanoparticles encoding the CCHFV glycoproteins or N demonstrated strong, protective cellular immune responses [
119]. Adoptive transfer of serum Abs and T cells from mice immunized with a modified vaccinia Ankara virus vector expressing the CCHFV glycoprotein protected recipient mice against lethal challenge [
121] . Depletion of either CD4+ or CD8+ T cells significantly increased mortality in infected mice, underscoring the essential role for these cell types in protection against severe disease [
122]. Finally, recent findings have also highlighted a crucial role for CD8+ T cells in efficiently controlling acute infection in wild type mice, rapidly acquiring CCHFV-specific antiviral effector functions, including the production of antiviral cytokines [
55].
While early studies in CCHFV patients suggest that cellular immunity enhances survival during acute infection [
54,
123], the exact mechanisms by which T cells contribute to survival remain to be investigated. Adaptive immune responses to other nairoviruses, especially NSV, are also underexplored. Hazara virus (HAZV), closely related to CCHFV, has served as a biosafety level 2 (BSL-2) surrogate model for CCHFV research, facilitating research without the requirement and constraints of a high-containment BSL-4 environment. Studies on HAZV have helped reveal important insights into CCHFV immunopathogenesis; however, ex vivo and in vivo studies exploring T cell responses against HAZV require further investigation [
124,
125]. Further understanding the mechanisms of viral clearance mediated by T cells will be important for designing effective vaccines against CCHFV and other nairoviruses.
Arenaviridae
The
Arenaviridae family currently encompasses five genera and 74 virus species with the capability of causing infections in diverse hosts. Mammarenaviruses, which include pathogens typically not infecting mammals beyond their primary reservoir hosts, post a threat to humans through direct contact with infected rodents, their droppings or urine, ingestion of contaminated food, or inhalation of aerosolized droplets from contaminated rodent excreta, secreta, or body parts [
126]. Human diseases caused by mammarenaviruses include Lassa fever (LF), caused by Lassa virus (LASV) in Western Africa. Lujo virus (LUJV) has also recently caused a small but severe outbreak in Southern Africa [
127]. Other mammarenaviruses, including Junin (JUNV), Machupo (MACV), Guanarito (GTOV), Sabia (SBAV), and Chapare (CHAPV) viruses cause human disease most often associated with hemorrhagic syndromes throughout South America.
Arenaviridae also includes lymphocytic choriomeningitis virus (LCMV), a well-studied virus that has facilitated many advances in the fields of virology and immunology, although not a major focus of this review [
128].
Beyond studies involving LCMV, T cell responses against LASV and other mammarenaviruses have also been characterized [
56,
57,
58,
129]. These studies focused on identifying immunogenic epitopes against entire proteomes for several mammarenaviruses strains (LASV, LUJV, CHAPV, JUNV, MACV, GTOV and SABV), with the goal of identifying conserved epitopes among the family [
56,
129,
130,
131]. Immunoinformatic analysis identified several highly immunogenic epitopes, mostly all located in conserved regions of GPC and N [
56]. Ex vivo stimulation of LF survivor cells narrowed down the panel of immunodominant epitopes to 12 CD8+ T cell positive epitopes within GPC and N which induced broad peptide-specific T cell responses, supported by predictive HLA-binding algorithms (
Table 1) [
57,
58]. Further, 4 immunodominant CD4+ T cell epitopes, which are highly conserved between Old and New World arenaviruses, were identified and mainly localized to a short stretch of 13 amino acids located in the N-terminal part of GP2
(289-301) (
Table 1) [
59]. Another study also showed strong human memory CD4+ T cell responses against N during LASV infection [
60]. In mice, CD4+ T cells specific to GPC
(403-417) of LASV can mediate cross-protective immunity to LCMV infection [
61]. Notably, the immunogenicity of GPC peptide candidates was evaluated in HLA-A*0201 mice which were protected against challenge with a recombinant vaccinia virus that expressed the LASV GPC [
132,
133].
In human LASV infection, T cells play a major role in controlling acute infection, as patients recover in the absence of a measurable nAb response [
134,
135] Furthermore, treatment with immune plasma did not protect LF patients, strongly suggesting a critical role of cell-mediated immunity against LASV infection in humans [
136]. Survival and LASV clearance in humans correlate with robust virus-specific CD4+ and CD8+ T cell responses during acute stages, coupled with elevated early IFN levels [
57]. In contrast, severe LF cases are associated with weak LASV-specific T cell responses and non-specific T cell activation [
137,
138]. Currently, our understanding of CD4+ T cell response to LASV infection is limited to observations of LASV-specific CD4
+ T cells in convalescent patients [
59,
60].
Our knowledge about JUNV-specific T cell responses is restricted to a few mouse studies, which implicated T cells in clearance of virus from infected organs and their correlation with disease severity [
139,
140]. The precise roles of CD4+ and CD8+ T cells, along with their epitope targets, remain unknown. Further investigations will help improve our understanding of the immunopathogenesis of JUNV and other arenavirus infections. Given that T cells play a protective role during infections with arenavirus, even in the absence of nAb responses, cross-protective T vaccines should be a major focus of future vaccine design and testing.