Abstract
Lassa virus (LASV), Junin virus (JUNV), and several other members of the Arenaviridae family are capable of zoonotic transfer to humans and induction of severe viral hemorrhagic fevers. Despite the importance of arenaviruses as potential pandemic pathogens, numerous gaps exist in scientific knowledge pertaining to this diverse family, including gaps in understanding replication, immunosuppression, receptor usage, and elicitation of neutralizing antibody responses, that in turn complicates development of medical countermeasures. A further challenge to the development of medical countermeasures for arenaviruses is the requirement for use of animal models at high levels of biocontainment, where each model has distinct advantages and limitations depending on, availability of space, animals species-specific reagents, and most importantly the ability of the model to faithfully recapitulate human disease. Designation of LASV and JUNV as prototype pathogens can facilitate progress in addressing the public health challenges posed by members of this important virus family.
Keywords: arenaviruses, knowledge gaps, animal models, countermeasures, prototype pathogen
Several members of the Arenaviridae family (order, Bunyavirales), including Lassa virus (LASV), Lujo virus (LUJV), Junin virus (JUNV), Machupo virus (MACV), Guanarito virus (GTOV), and Chapare virus (CHAPV), are capable of infecting humans [1, 2]. Infection by these viruses can lead to severe viral hemorrhagic fevers (VHFs) in a subset of individuals. Each are classified as biosafety level 4 (BSL-4) agents; however, JUNV may be used at BSL-3 if investigators are vaccinated. LASV is endemic in Western Africa and, after dengue virus, is the most common hemorrhagic fever virus imported into Europe and the Americas. Numerous other arenaviruses are endemic to the United States, although among them only Whitewater Arroyo virus has been isolated from cases of fatal VHF [3]. Here, we review the current state of knowledge about arenaviruses to identify gaps that hinder development of countermeasures.
TAXONOMY
The Arenaviridae family includes viruses that infect rodents, bats, shrews, snakes, and fish as well as humans. The family is comprised of 4 genera, each recognized as distinct, well-supported groups in phylogenies estimated based on the RNA-dependent RNA polymerase (L) protein (Figure 1 and Table 1). Old world (OW) and new world (NW) arenaviruses of mammals are placed taxonomically in the genus Mammarenavirus. These complexes were initially defined based on both serologic classification and geographic origin, but they are also clearly supported at the phylogenetic level [4]. OW mammarenaviruses are found in African rodents from the family Muridae [5–7], and in Asian rodents from various Rattus and Mus species as well as shrews (Suncus murinus) [8–10]. OW arenaviruses do not form clear phylogenetic clades. However, 4 established LASV lineages are broadly distributed across West Africa, with 3 additional proposed lineages recently emerged [11]. Lymphocytic choriomeningitis virus (LCMV), an OW mammarenavirus that is frequently used to model antiviral immune responses, is distributed worldwide in common house mice, Mus domesticus and Mus musculus [1].
Figure 1.
Phylogenetic relationships among species from the Arenaviridae family. Maximum-likelihood tree for L protein sequences representative of the genetic diversity within the Arenaviridae family. The phylogeny was inferred with the IQ-TREE software, using the LG + G4 model. Branch support was assessed using the ultrafast bootstrap approximation with 1000 replicates. Nodes with branch support >95% are indicated with circles. Tree leaves are according to the main groups within the family, namely the genera Hartmanivirus, Antennavirus, and Reptarenavirus, as well the new world complex and old world complex of the Mammarenavirus genus. These main groups are also shown by rectangles on the right part of the plot. For the new world complex, the main clades are indicated by their corresponding letters (A to D). For the Lassa virus species, lineages are indicated as part of the sequence name (*indicates that the lineage assignment is tentative).
Table 1.
Characteristics of Selected Arenaviruses
Taxonomic Position | Virus Common Name | Reservoir | Distribution | Disease | Vaccines | Therapeutics |
---|---|---|---|---|---|---|
Old world mammarenaviruses | Lassa virus | Multimammate mouse (Mastomys natalensis) and others | West Africa | In humans: Lassa fever | Multiple vaccine candidates in development | Small molecule drugs and a monoclonal antibody combination in development |
Lujo virus | Unknown | In humans: Lujo HF | None | None | ||
Lymphocytic choriomeningitis virus | House mice (Mus musculus and Mus domesticus) | Worldwide | In humans: influenza-like illness; aseptic meningitis; meningoencephalitis; congenital microencephaly | None | None | |
New world mammarenaviruses | Junin virus | House (Mus musculus), Vesper (Calomys callosus) and grass mice (Akodon azarae) | Argentina | In humans: Argentine HF | Candid-1 live attenuated vaccine | Passive survivor plasma; monoclonal antibodies in development |
Machupo virus | Vesper mouse | Bolivia | In humans: Bolivian HF | None | None | |
Guanarito virus | Alston's cotton rat (Sigmodon alstoni) | Venezuela | In humans: Venezuelan HF | None | None | |
Sabiá virus | Unknown | Brazil | In humans: São Paulo HF | None | None | |
Chapare virus | Pygmy rice rats (Oligoryzomys microtis) | Bolivia | In humans: Chapare HF | None | None | |
Whitewater Arroyo virus | Wood rats (Neotoma spp.) | Southwest United States | In humans: California HF | None | None | |
Wōnlnzhōnlu virus | Rats (Rattus spp.), mice (Mus spp.), and shrews (Suncus spp.) | China, Cambodia, Thailand | In humans: fever and respiratory symptoms | None | None | |
Hartmaniviruses | Haartman Institute snake virus 1 | Captive boas (Booidea) and pythons (Pythonidae) | Worldwide | None observed | None | None |
Reptarenaviruses | California Academy of Sciences virus | Captive boas and pythons | Worldwide | In snakes: Boid inclusion body disease | None | None |
Antennaviruses | Wēnlng frogfish arenaviruses | Frogfish (Antennarius striatus) | China (likely more widespread) | Unknown | None | None |
Salmon pescarenavirus | Wild and farmed Chinook (Oncorhynchus tshawytscha) and sockeye (Oncorhynchus nerka) salmon | Northeast Pacific Ocean | In salmon: spleen and liver inflammation; tubule necrosis and hyperplasia in the kidney | None | None |
Abbreviation: HF, hemorrhagic fever.
NW mammarenaviruses infect rodents of the family Cricetidae [3, 12], with the exception of Tacaribe virus that infects artibeus bats [13] (Figure 1 and Table 1). Species from the NW complex form 4 well-defined clades [14]. Clades A–C are distributed across Latin America and the Caribbean, while all current representatives of clade D have been found in North America. NW mammarenaviruses in North America are maintained in reservoir rodents from the Cricetidae subfamily Neotominae, whereas Latin American and Caribbean mammarenaviruses are maintained in rodents from the subfamily Sigmodontinae [15, 16]. Arenaviruses of snakes [1, 17, 18] are classified in 2 genera, Reptarenavirus and Hartmanivirus. Arenaviruses first detected in frogfish (Antennarius striatus) [19], and later in salmon [20], represent a new genus, Antennavirus [21].
Knowledge Gap
The extent of arenavirus diversity is unknown. The Arenaviridae family likely extends to other yet uncharacterized vertebrate hosts, including additional species of mammals, reptiles, and fish. Because of the potential for spillover to humans it would be prudent to further characterize NW mammarenavirus diversity, which is much less well understood than for OW viruses. Screening of rodents, shrews, bats, and other potential arenavirus reservoirs by metagenomic sequencing should be utilized to elucidate the as yet undersampled diversity of NW mammarenaviruses.
MOLECULAR VIROLOGY
Arenaviruses have pleomorphic enveloped virions ranging in size from 80 to 150 nm (Figure 2). Electron-dense granules, considered to be host-derived ribosomes, give virions their “sandy” granular appearance in electron micrographs that led to the family name (Latin arenosus: sandy) (Figure 2A). Genetic differences are readily apparent when comparing mammalian, reptilian, and fish arenaviruses (Figure 2B). Mammarenaviruses, reptarenaviruses, and hartmaniviruses have bisegmented genomes [22]. The small segment is ambisense and encodes the glycoprotein complex (GPC) and the nucleocapsid protein (NP). The large segment encodes the RNA-dependent RNA polymerase (L) and, in mammarenaviruses and reptarenaviruses in which the large segment is ambisense, a zinc-binding or matrix protein (Z). Hartmaniviruses and antennaviruses do not encode a Z protein [23]. Antennaviruses contain 3 genome segments [19], the largest of which encodes L; GPC and another small protein of unknown function are encoded by another segment with the NP homolog encoded separately by the third genome segment.
Figure 2.
Arenavirus proteins and genomes. A, The arenavirus virion is enveloped and contains 2 or 3 RNA subunits, plus cellular ribosomes. B, Genome structures of the 4 genuses in the Arenaviridae family. X is a protein of unknown function encoded by antennaviruses. Plus symbol indicates that the gene is positive polarity. Negative symbol indicates the gene is negative polarity. Lines indicate proteins with significant sequence similarity.
GPC of mammarenaviruses, hartmaniviruses, and antennaviruses include an unusual long stable signal peptide (SSP) that is retained in the virion as part of the GPC. The receptor-binding glycoprotein 1 subunit (GP1) and a transmembrane fusion subunit GP2 are arranged on the virion surface as a trimer of dimers [24]. The glycoprotein trimer bears a thick glycan shield [25], and recognition by neutralizing antibodies is rare [24, 26]. Reptarenavirus glycoproteins are distinct from other arenavirus glycoproteins, but are related to glycoproteins of filoviruses and retroviruses [27]. Reptarenaviruses do not encode a SSP.
Due to a transcriptionally regulated requirement for a threshold amount of NP, this protein is the most abundantly produced protein during arenaviral infections. NP is divided into N- and C-terminal domains (Figure 3). The N-terminal half of NP adopts a clam-shell shape in which N- and C-terminal subdomains encase 8 bases of single-stranded RNA (ssRNA) [28] (Figure 3A). The C-terminal domain functions as a 3′–5′ exoribonuclease that specifically and rapidly digests double-stranded RNA (dsRNA) [29–31], a key inducer of the interferon system and innate immunity (Figure 3B). The exoribonuclease appears to play a key role in LASV-induced immunosuppression. In the absence of RNA, NP forms a trimer with N- and C-terminal domains oriented in a head-to-tail fashion in an RNA-free trimeric ring of NP monomers [32]. This oligomeric arrangement of NP is incompatible with RNA binding. This assembly could represent a conformation-driven chaperoning strategy of the unbound NP. Indeed, binding of RNA requires the opening of an α-helical gate (Figure 3A) to expose the ssRNA pocket [28]. Relocation of this gate would prevent trimerization.
Figure 3.
Structure and RNA binding of Lassa virus nucleoprotein (LASV NP). A, Cartoon representation of the N-terminal domain of LASV NP showing the deep groove through which the single-stranded RNA (ssRNA) channels. Conformational changes open and close the RNA-binding pocket. α-helix6 (α6) is positioned on top of the pocket, preventing RNA from binding. When α6 shifts away from the pocket as shown, the RNA gate opens to accommodate ssRNA. B, Cartoon representation of LASV NPΔ340, the C-terminal, immunosuppressive domain of LASV NP. Double-strand RNA (dsRNA), a potent interferon inducer, is degraded by the exonuclease activity of NP. Residues with similarity to the DEDD superfamily of exonucleases show coordination of a magnesium (Mg) ion. At the entrance to the active site of the exonuclease are several positively charged amino acids. Coordination of a zinc (Zn) ion is indicated.
The matrix protein Z of mammarenaviruses occurs in monomeric and oligomeric forms [33]. In mammarenaviruses, the glycine at position 2 is myristoylated, which is important for association with the plasma membrane, and the really interesting new gene (RING) domain of Z contains 2 zinc-binding domains [33, 34]. In reptarenaviruses, Z also has a RING domain, but unlike mammarenaviruses, the reptarenavirus Z contains a transmembrane domain [35]. For the mammarenaviruses, several recent studies have described the binding of monomeric Z to L where the RNA product exits [36], and locking of the different domains of L together [37], suggesting a role of Z in regulating the polymerase [36–39].
Knowledge Gaps
Z is known to negatively regulate L, but the concentration dependence is not sufficiently understood. The conformational and functional switch between the function of Z in control of L in the cytosol, and function of Z as the viral matrix protein at the plasma membrane also remain unclear. The dodecameric ring form of Z has no known function. Furthermore, the role of the Z myristol group in the soluble Z–L is unknown including whether Z traffics to the membrane separately from L, the role of the oligomer, and what conformational changes permit and then destabilize the Z–L interaction allowing L to begin RNA synthesis. Further elucidation of the roles of Z and L in regulating viral RNA synthesis could potentially facilitate development of arenavirus small molecule drugs targeting this process.
Mechanisms of immunosuppression by Z and other arenaviruses proteins are incompletely described. Filling this gap may allow development of medical countermeasures that block this process.
Assembly mechanisms of hartmaniviruses and antennaviruses that do not encode a Z protein are unknown.
ENTRY AND REPLICATION
Cell entry of LASV and other arenaviruses is a multistep procedure. Several host membrane proteins can serve as cell surface receptors/factors for the prefusion (virion) form of GPC. The major and most efficient cell surface receptor for LASV entry is matriglycan on α-dystroglycan (α-DG) [40–45]. Clade C NW mammarenavirses also utilize α-DG for entry. In contrast, the major cell surface receptor of clade B NW mammarenaviruses is the transferrin receptor. No receptors have yet been identified for clade A and D mammarenaviruses.
After binding to surface receptors, LASV virions are internalized via endocytosis. Prefusion GPC responds to the acidic endosomal pH inducing a conformational shift in prefusion GPC termed “GPC priming,” and dissociation from α-DG. Primed GPC binds to the endosomal receptor, lysosomal-associated membrane protein 1 (LAMP-1) [46, 47]. Following LAMP-1 binding, GPC undergoes additional conformational changes that mediate virus-endosomal membrane fusion and LASV genome release into the cytosol. LAMP-1 is not absolutely required for GPC-mediated membrane fusion in very late endosomes, but if it is not present, entry is inefficient [48, 49]. Like LASV, LCMV and its close relatives were recently found to switch receptors from α-DG to CD164 in the endosome [50]. Similarly, a genetic screen implicates the endosomal resident CD63 in LUJV entry [51].
After the viral RNA is unpacked, RNA replication and transcription take place in the cytoplasm. In the case of the bisegmented arenaviruses, primary transcription of the S and L genomic RNAs produces mRNAs that encode NP and L, respectively. S and L RNA genomes serve as templates for antigenomic RNAs, which are the templates for GPC and M mRNAs as well as for the full-length progeny genomes. A stem-loop structure within the intergenomic region terminates transcription from genomes and antigenomes. Z functions as a negative regulator of L. Arenaviruses use a cap-snatching strategy involving the endonuclease activity of L, which is required for effective translation from cellular mRNAs [52]. The GPC precursor is tracked from the endoplasmic reticulum to the Golgi, where it is heavily N-glycosidated and processed by cellular proteases (SPase, SKI1/SP1) into its mature form comprising noncovalently linked GP1, GP2, and SSP. NP encases the viral genome via its N-terminal domain. Cleaved glycoproteins are incorporated into the virion envelope when N, M, and L and the genomic RNA are assembled. The virus then buds and is released at the cell surface membrane.
Knowledge Gaps
While the molecular structures of proteins of LASV and certain NW arenaviruses, including MACV and JUNV, have been determined, there is limited structural information for proteins of other arenaviruses, particularly those outside the Mammarenavirus genus. Structural information for GPC coupled with identification of key epitopes can inform development of vaccines and immunotherapeutics. Structural information for other arenaviruses proteins, such as L, could facilitate development of small-molecule drugs. Further clarity on functional significance of each protein during replication and in the context of infection are also needed.
Z appears sufficient for virus budding in those arenaviruses that encode a Z. The mechanisms by which Z mediates virion formation are unknown. How arenaviruses lacking Z assembe virion particles and egress from the cell is unclear.
The exonuclease domain of NP rapidly mediates dsRNA digestion, and this plays a role in blocking innate immune sensing in cell culture. Furthermore, exonuclease knockout mutations are still impaired in interferon-knockout cells, suggesting that the exonuclease function may play an additional fundamental role in viral replication and propagation or in proofreading. Improved understanding of the role of exonuclease domain of NP in vivo could potentially be exploited for developing medical countermeasures (MCM).
The function of the RNA-free NP trimer is unknown. It may be an artifact of recombinant overexpression. Another possibility is that it represents an “NP0” in which trimerization of NP performs the equivalent function of the P protein of other negative-strand viruses, which arenaviruses do not explicitly encode. Identification of the specific role of the trimer may guide design of inhibitors targeting arenavirus replication.
There is an incomplete understanding of entry for NW mammarenaviruses. It is unknown whether the receptor switch utilized by LASV, LCMV, and LCMV-like viruses is common to other OW mammarenaviruses.
EPIDEMIOLOGY
Rural areas of the West African nations of Nigeria, Sierra Leone, Liberia, and Guinea are endemic for Lassa fever [53]. Mounting evidence points to expansion of the Lassa fever zone to include Burkina Faso, Mali, Togo, Benin, Senegal, and Côte d’Ivoire. The true incidence of Lassa fever has not been determined, in part because of the lack of available diagnostic assays. Most LASV-infected persons in West Africa are likely never seen by health professionals due to limited access to medical care. The initial symptoms of Lassa fever are like other febrile illnesses, leading to misdiagnoses of other common endemic illnesses such as malaria. Extrapolations from prior limited serological studies suggested that there may be as many as 500 000 LASV infections and 5000 deaths per year in West Africa [54, 55]. The development of recombinant antigen enzyme-linked immunosorbent assay (ELISA) has recently enabled large-scale serological surveys that are underway at the time of this writing in several West African countries. Ongoing seroprevalence studies of LASV in West Africa are essential for conducting human clinical trials of MCM such as vaccines or therapeutics.
Argentine hemorrhagic fever was first described in 1955 [56]. It manifests as a number of catastrophic vascular, renal, and neurological symptoms and has a mortality rate of 15%–30% in untreated individuals. The etiologic agent of Argentine hemorrhagic fever is an arenavirus, JUNV. Before the development of a vaccine in 1992, there were hundreds of cases of Argentine hemorrhagic fever each year [57].
Knowledge Gaps
Arenavirus reverse genetics systems exist, but need development and support. These may be critical in times of an outbreak of a novel arenavirus to quickly begin working safely and expeditiously on MCM, such as diagnostics, vaccine, and drug development, as soon as genetic information is available.
Arenaviruses tend to be geographically restricted, reflecting the range of their reservoir hosts. JUNV, MACV, GTOV, Sabia virus (SABV), and CHAPV, cause Argentine, Bolivian, Venezuelan, Brazilian, and Chapare hemorrhagic fevers, respectively. The prevalence of Argentine hemorrhagic fever has been described; however, the incidence and prevalence of other South American arenaviruses is largely unknown.
Importation of samples for characterization of arenaviruses under high containment has challenges, including export/import hurdles and regulations involving select agents. Fortification of clinical and basic research infrastructure, particularly in areas known to be endemic for arenaviruses, is needed to circumvent these challenges. Another measure can be establishment and support of symmetrical international research collaborations with prenegotiation of import and export licenses.
It is unclear if all isolates of OW and NW arenaviruses, including those that have been demonstrated to cause VHFs, cause serious disease. Nonlethal strains of mammarenaviruses may exist that could inform studies of disease pathogenesis that could be modelled in experimental animals.
ECOLOGY
With no approved vaccines or therapeutics available, Lassa fever is among the most consequential rodent-borne virus infection in terms of morbidity, mortality, and societal costs. Individuals living in endemic hotspots of Lassa fever have recurrent exposure to LASV via spillover from the primary host reservoir Mastomys natalensis especially in or near human habitations. Additional rodents, including Rattus sp., the African wood mouse Hylomyscus pamfi, and the Guinean multimammate mouse Mastomys erythroleucus, also appear to be involved in LASV transmission to humans. Individuals contract LASV through inhalation of aerosolized urine in droplets or dust particles, as well as via ingestion of food or water or contact with fomites contaminated with rodent urine or droppings. Knowledge regarding rodent habitats, reproduction and fecundity, movement patterns, and spatial preferences are essential for implementing effective preventative measures against Lassa fever. The efficacy of rodent abatement practices, including the efficacy of feline and chemical rodenticides, is of equivalent importance to drive stringent rodent control.
Like LASV, the detected range of JUNV incidence is not in perfect alignment within the geographic range of its primary host, Calomys musculinus [58, 59]. The first outbreaks of Argentine hemorrhagic fever were primarily in the humid pampas of central Argentina, 200–300 km west of Buenos Aires. Since then, the endemic area has expanded northwards and westwards from its historical foci to its present extent of approximately 150 000 km2 [59, 60]. JUNV is also present in animals outside the historic Argentine hemorrhagic fever endemic zone [57]; however, the geographic range of JUNV has not expanded to the full range in which the host species are found. Since the discovery of JUNV, several other South American mammarenaviruses causing severe hemorrhagic fevers, including MACV (Bolivia), SABV (Brazil), GTOV (Venezuela), and most recently CHAPV (Bolivia), were discovered associated with sporadic outbreaks of severe human disease [61]. The incidence of exposure to the human population by these viruses is largely unknown as only severe cases are the most likely to be reported.
Knowledge Gaps
Despite M. natalensis being broadly distributed across sub-Saharan Africa, Lassa fever is only found in West Africa. Factors that contribute to the geographical distribution of LASV are poorly described. The features of M. natalensis populations that carry LASV and those that do not should be determined.
Known arenaviruses, including LASV, may have a broader host range than currently documented. Other rodents could be involved as transient or intermediate vectors. Likewise, the ecology and geographic distribution of NW mammarennaviruses has not been well elucidated. A better understanding of arenavirus ecology, including several rodent species that serve as reservoirs or intermediates of LASV transmission to humans are needed, can inform public health interventions.
PATHOGENESIS
Infection with LASV can result in a range of outcomes from a mild or asymptomatic infection to a severe life-threatening illness. Signs and symptoms of Lassa fever occur 1–3 weeks after exposure and are highly variable. After 4–7 days of mild illness, an estimated 20% of infected individuals develop symptoms of a severe VHF. Sore throat, vomiting, and coughing are common, but each case presentation is distinct [54, 62, 63]. Around 40% of Lassa fever patients experience bleeding from the nose, mouth, other orifices, and mucosal surface, which confers a poor prognosis. Death from Lassa fever generally occurs between 10 and 14 days of symptom onset and is attributed to diminished effective circulating volume, shock, and multiorgan system failure [63]. Infection is approximately 90% lethal to women in the third trimester of pregnancy. Neurological problems have also been described following LASV infection, including tremors, and encephalitis. Temporary or permanent unilateral or bilateral deafness has been reported to occur in approximately 30% of patients with Lassa fever [64, 65].
Mechanisms of pathogenesis by LASV and other arenaviruses are incompletely understood, but are thought to include direct cytopathic effects by viral proteins, including increase in vascular leakage. Immunopathogenesis, including induction of a cytokine storm, also appears to be involved. Long-term sequelae, including neurological, immune, and renal defects, have been described in survivors and may involve the induction of autoimmunity. The risk of recrudescence has yet to be ruled out.
Knowledge Gaps
The morbidity burden of LASV in endemic regions is unclear. Case fatality rates are based on patients presenting to health care facilities which overrepresents severe LASV infections, and do not include asymptomatic or mild infections that likely predominate in the wider population.
Factors that determine virulence, severity of disease, and outcome of arenavirus diseases are unknown. There are likely a multitude of factors, including human and viral genetic differences. Different lineages of LASV have different case fatality rates, which could reflect these genetic factors, as well as differences in clinical management. It is possible that OW and NW mammaraenaviruses employ different pathogenic mechanisms.
Field-caught rodents that are carrying arenaviruses do not display obvious signs of illness. However, the pathogenesis of arenaviruses in reservoirs or intermediate species is not known. Infection studies of M. natalensis raised and bred in captivity could be informative in this regard.
While sometimes present in healthy snakes, reptarenaviruses are the causative agent of inclusion body disease, which spreads among boid snakes (boas and pythons) in captivity [27, 66–70]. Hartmaniviruses have not been shown to cause inclusion body disease [71].
Antennaviruses of salmon, which have been isolated from dead or moribund aquaculture salmon as well as live-sampled wild salmon, appear to be pathogenic. Mechanisms of pathogenesis of arenaviruses other than the mammarenaviruses have not been determined.
NATURAL IMMUNITY AND IMMUNOEVASION
Protective immunity has been correlated with cellular immune responses to LASV rather than humoral responses. Responses to both GPC and NP have been detected. Some T cell epitopes are lineage-restricted, and others are pan-lineage. There is significant cross-reactivity of cellular responses in humans across LASV lineages. Putative conserved T cell epitopes have been identified. While infected humans and experimental animals such as nonhuman primates (NHP) develop strong antibody responses to LASV, those antibodies are often directed to epitopes not associated with viral clearance such as nonneutralizing epitopes or linear epitopes exposed only in the postfusion conformation. The LASV GPC trimer bears 33 N-linked glycans that form a denser shield relative to other human viruses; only the human immunodeficiency virus (HIV-1) has a heavier glycan shield [72]. In addition, LASV infection and overexpression of GPC in immunogen production leave a large proportion of uncleaved GPC molecules that cannot mediate membrane fusion [73]. Consequently, this glycan shield together with GPC in nonprefusion structures is thought to pose a substantial barrier to elicitation of neutralizing antibodies. LASV neutralizing antibodies, if detected at all, are usually only present after recovery. Neutralizing epitopes require prefusion GP and are usually quaternary. Significant cross-reactivity of humoral responses in humans neutralizing antibody titers continue to rise even several months after convalescence has been established, which may indicate constant stimulation of B cells due to low levels of virus persistence.
Knowledge Gaps
While convalescent plasma is the standard for treatment of Argentine hemorrhagic fever caused by JUNV, convalescent plasma has not been shown to be an effective treatment for LASV infection. The differences in natural immunity that account for this difference have not been elucidated, but likely may relate to the types of neutralizing antibodies elicited (see below).
It remains to be determined whether it will be possible to design vaccines that effectively elicit neutralizing antibodies such as those that manifest during the late stages of Lassa fever convalescence.
Natural immunity to LUJV and to NW mammarenaviruses has not been extensively characterized.
It is unknown whether T cell immunity induced by natural infection with mammarenaviruses induces cross-protective immunity across lineages. NP and potentially other arenavirus proteins may induce protective T cell–mediated immunity.
Some arenaviruses proteins are known to have immunosuppressive properties, but the mechanisms are incompletely understood.
For GPC, we now have structures of prefusion GPC in complex with a few neutralizing antibodies, and we understand why neutralizing antibodies may be difficult to elicit. Additional work is needed to turn these predictions into vaccines to determine how a neutralizing antibody response could be elicited by vaccination.
ANIMAL MODELS
Animal models are required to investigate arenavirus-induced disease pathogenesis and for developing therapeutics and vaccines [74, 75]. Inbred Strain 13 guinea pigs infected with wild-type LASV (strain Josiah) show a fatality rate close to 100%. These animals also show a high rate of mortality when infected with LUJV and several NW mammarenaviruses. A limitation to the use of inbred strain 13 guinea pigs is the lack of commercial vendors—researchers that wish to use them require private colony support. Outbred (Hartley) guinea pigs are commercially available. These Hartley guinea pigs, however, show a LASV fatality rate of only approximately 30% [76–78], although a guinea pig adapted LASV has been developed with near 100% lethality in outbred guinea pigs [79]. Guinea pig-adapted strains of LCMV and certain NW mammarenaviruses that can be used at BSL-3 have also been developed, although the outcome of infection is strain dependent. While these models have demonstrated fantastic predictive efficacy for success of vaccines and therapeutics in NHP, they are of limited value for studying host responses due to limited reagents available for immunological characterization in guinea pigs.
Wild-type laboratory mice are not highly permissive to infection by LASV and most NW mammarenaviruses, and mice fail to develop quantifiable pathologies. Conversely, certain strains of immunocompromised or knock-out mice are susceptible. Interferon-α receptor knock-out (IFNAR−/−), human/mouse-chimeric HLA-A2.1 (humanized HHD), chimeric IFNA−/−B6, CBA, and STAT deficient mice (STAT1−/− mice) show varying manifestations of LASV disease, ranging from semi- to fully lethal [74, 75]. These mice have been used to study sensorineural hearing loss after recovery from LASV [80]. Immunocompetent 3-week-old transgenic mice expressing hTfR1 are highly susceptible to JUNV infection and develop lethal disease [81]. The development of this novel small-animal model provides a valuable tool for the study of JUNV pathogenesis and a platform for the evaluation of therapeutic interventions that interfere with virus binding to hTfR1 in vivo. Murine models of arenavirus infection have utility because many immunological reagents are available; however, the impact of each model’s immunological deficiency should be appropriately accounted for.
While rhesus macaques, common marmosets, and squirrel monkeys have been described as LASV models, the most frequently used NHP model is the cynomolgus macaque due to a more consistent susceptibility. Disease manifestations in all NHP models closely mimic that of severe Lassa fever and are likely the most predictive for MCM efficacy in humans. Disease severity in NHP models is understood to be dependent on LASV strain and dose [74, 82, 83]. NHP models benefit from the availability of many useful immunological reagents due to cross-reactivity of monkey antigens with human antigens and heavy investment from the biomedical field. Survival models tied to hearing loss observed in humans have been described [84], but highlight the challenges of performing survival studies with NHP in high containment where studies often cannot go over 45 days postchallenge. Nonetheless, the requirement for BSL-4 facilities, where space and study prioritization factor heavily into study implementation, have been an ongoing limitation. Perhaps most concerning, due to shifts in global animal trade caused by the COVID-19 pandemic, NHP resources are becoming increasingly scarce, which has further compounded difficulties in carrying out pathogenesis studies and critical evaluations of MCMs [85].
Knowledge Gaps
Route of challenge may impact disease course and thus MCM efficacy. Most infection models employ an intraperitoneal (rodent) or intramuscular/subcutaneous (NHP) challenge route; yet natural infection (exposure to rodent excreta) likely is the result of mucosal exposure in the host. While limited small-particle aerosol models do exist in the context of modeling deliberate release, there are few if any data on more traditional mucosal challenge models.
Some of the first detections of LCMV infections in human were in transplant patients and areas of Africa where coinfections with HIV and malaria are common. MCM evaluation and pathogenesis studies for LCMV and other arenavirus infections should also be performed in immunocompromised populations.
There exist only limited data on survival models of OW or NW mammarenaviruses.
Not all isolates are lethal in NHP despite being from human fatal cases. Mechanisms that determine virulence are unclear.
ALTERNATIVES TO WORK WITH BSL-4 PATHOGENS
Handling most arenaviruses of public health priority requires high containment laboratories. Potential solutions to high containment work include research with virus-like particle (VLP) systems. However, VLPs provide an incomplete picture of events of infection after entry or host responses to productive infection. Pseudotyped viruses are also useful for study of entry and related MCM development, but not reflective of the pleomorphic shape of most arenavirus and do not undergo postentry replication steps. Surrogate virus infection models (ie, Pichinde virus in hamsters, LCMV in mice and guinea pig) may be useful in predicting viral entry, but lack key pathogenic features only found in authentic isolates. Surrogate viruses also have limited value where antigen is central to countermeasure efficacy or understanding pathogenesis.
Knowledge Gap
1. Surrogate virus systems, particularly those that can utilize murine models at BSL-2, have not been a research priority and are underdeveloped.
DIAGNOSTICS
Lassa fever is difficult to recognize on clinical grounds alone, especially in the early stages. Prompt laboratory diagnosis is therefore essential for treatment to begin as early as possible. While still limited to some central laboratories, Lassa testing has become more readily available in endemic regions of West Africa. Laboratory-made and commercial polymerase chain reaction (PCR) assays are available. Likewise, LASV antigen-capture and IgM- and IgG-capture ELISA using recombinant LASV proteins have been produced and characterized [86]. Immunoglobulin M antibodies appear not to be a reliable indicator of recent LASV infection, as this class of anti-LASV antibody can persist for months or years after infection [87]. A lateral flow immunoassay for LASV antigen can reliably detect LASV antigen from a drop of blood of patients obtained by a simple fingerstick. In principle, rapid tests could be disseminated to outlying clinics and laboratories and reliably and safely performed by persons with limited laboratory training.
Knowledge Gaps
Diagnostics for NW mammarenaviruses are not generally available.
The extent of antigenic cross-reactivity between proteins of LASV and proteins of other OW arenaviruses in Africa remains to be determined. Exposure to non-LASV arenaviruses could influence seroreactivity.
IMMUNOTHERAPEUTICS
Humoral immune responses play a minor role, if any, in recovery from LASV infection. Nevertheless, immunotherapy appears to provide a significant therapeutic benefit. It is well established that convalescent plasma from survivors of Argentine hemorrhagic fever, caused by JUNV, can increase survival of people with acute infection. Convalescent plasma is the only approved treatment for any arenavirus infection [88, 89]. Plasma from human Lassa fever survivors has also been used to treat Lassa fever in Cynomolgus macaques [90, 91]. In contrast, convalescent plasma was less effective than ribavirin in treating acute Lassa fever [92]. Although neutralizing antibodies against LASV GPC are rare, 16 neutralizing human monoclonal antibodies (mAbs) from survivors, some of whom had long-term, multiple LASV exposure, have been cloned and expressed. Three of these antibodies, 8.9F, 12.1F, and 37.2D, have potent and broad neutralization activities against multiple LASV lineages [26]. A monoclonal antibody cocktail, termed arevirumab-3, comprising these 3 antibodies, provides complete protection in Cynomolgus macaque models, even when delivered in late-stage Lassa fever (8 days postinfection with death typically occurring on day 11) and at low doses [93, 94]. Arevirumab-3 also protects Cynomolgus macaques after challenge with LASV lineages II and III (unpublished, Cross et al). Work is underway to validate this treatment against other LASV lineages.
All neutralizing antibodies characterized to date target prefusion LASV GP and nearly all bind to quaternary epitopes that span the GP1 and GP2 subunits. These antibodies have various mechanisms of action, including likely competition with cellular entry factors and prevention of the LAMP1 receptor switch or fusion of viral and host cell membranes (Li, Hastie, and Saphire, unpublished observations). In contrast, thus far, all neutralizing antibodies targeting NW viruses bind to the GP1 subunit alone and all function by direct competition with TfR1. Furthermore, elicitation of neutralizing antibodies against JUNV is possible through vaccination with the GP1 subunit alone [95].
No evidence yet exists that NW arenaviruses employ receptor switching in the endosome, nor does structural analysis of several NW GP1 subunits suggest that conformational changes occur upon exposure to low pH or separation from GP2 [24]. Hence, the difference in epitope requirements for neutralizing antibodies may reflect the particular entry strategy employed by NW viruses versus LASV. Variations in the entry pathways may also be an underlying reason for the disparity in the effectiveness of convalescent plasma between the 2 groups of viruses.
Although neutralizing antibodies against NW arenaviruses appear easier to elicit, their breadth is limited. Of anti-JUNV antibodies identified to date, most are not cross-reactive against MACV [96] and none neutralize other clade A viruses [97]. The primary underlying cause of the narrow activity for these antibodies is high sequence variability in the receptor binding site of NW viruses. For example, MACV GP1 contains an insertion in a loop that interacts with TfR1 yet blocks binding by an anti-JUNV GP1 neutralizing antibody. Similarly, other NW arenaviruses have additional N-linked glycans that may occlude access by antibodies [98].
Therapeutic antibodies are available or in development for JUNV [99] and MACV [100]. All NW mammarenavirus neutralizing antibodies identified to date are against the GP1 subunit and block TfR1 binding. It appears that exposure to the GP1 subunit of NW mammarenaviruses alone is capable of neutralizing antibodies. As the GP1 subunit may be presented authentically, no matter the conformation of the GP2, it may be more straightforward for an infected or immunized person to develop anti-GP1 neutralizing antibodies. Furthermore, such antibodies may be easier to detect as the GP1 subunit in isolation is easier to make recombinantly and is not subject to the conformational heterogeneity of a complete GPC. The formal possibility remains that there could be other types of neutralizing antibodies against NW arenaviruses that involve GP2-containing epitopes. In the absence of well-formed GPC maintained in the prefusion state through the use of molecular clamp technology, such antibodies are difficult to identify for NW mammarenaviruses.
Knowledge Gaps
It is unclear whether the sequence variability in receptor-binding domains of NW mammarenaviruses is a barrier to cross-protective vaccines and/or Ab-based therapeutics. It remains to be determined if receptor use is related to the types of neutralizing antibodies elicited or to the antibody repertoire needed to generate a protective response.
It is unknown whether neutralizing antibodies against GP2-containing quaternary epitopes for OW or NW mammarenaviruses exist.
mAbs provide stunning late-stage and low-dose protection against LASV in NHPs. However, clinical trials of LASV and NW mammarenavirus mAbs in humans have not yet been conducted. Opportunities for more broadly cross-reactive neutralizing antibodies against the NW or OW arenaviruses may be available.
Beyond LASV, no human antibodies have yet been described for LCMV or LUJV. Some neutralizing antibodies from mice have been described for LCMV, but none for LUJV. Some neutralizing antibodies against LASV can neutralize LCMV, but none neutralize authentic LUJV (Cross, unpublished).
It is unclear whether similar antibodies to the GPC-specific antibodies against LASV exist for NW or other OW mammarenaviruses.
While immunotherapeutics are very promising, most arenaviruses have neurotropic tendencies. Monoclonal antibodies are unlikely to penetrate the blood-brain barrier. Options available to treat the infected central nervous system may include small molecules or nanobodies.
VACCINES
The live-attenuated Candid#1 strain of JUNV is the only approved arenavirus vaccine [101]. Candid#1 is licensed for use in Argentina; however, due in part to the possibility of genetic reversion to virulence of its single GP2 attenuating mutation, Candid#1 is not licensed in the United States [102]. Multiple platforms have been evaluated as potential Lassa fever vaccines, including a ML29 MOPV/LASV live reassortant (L segment from Mopeia virus, S segment from LASV) [103], recombinant vesicular stomatitis viruses in which the glycoprotein gene is replaced by the coding sequence for LASV GPC [104, 105], plasmid DNA encoding codon optimized GPC [106], recombinant measles virus [107], and a vaccinia virus recombinant vaccine (modified vaccina virus Ankara-virus-like particle) expressing LASV GPC + Z [108]. Additional vaccine candidates include a rabies virus recombinant expressing LASV GPC [109], adenovirus vectored vaccines [110, 111], LASV VLP [112, 113], and a virus replicon particle vaccine [114]. Important considerations for LASV vaccines stem from their use in Africa. They should be cost-effective, stable without extensive cold chain requirements, and require a minimal number of vaccinations. Efforts to develop vaccines for other arenaviruses beyond JUNV and LASV have been limited.
Knowledge Gaps
Most advanced experimental vaccines target GPC, but it is unclear whether other viral targets, such as N, can be exploited. While the majority of these platforms demonstrated a robust immune response in laboratory animals, many have failed to fully protect against lethal disease in relevant disease models. Both T and B cell responses may be required for optimal protection against OW or NW mammarenaviruses.
It is unclear whether vaccination by one lineage of LASV can protect against other lineages. Most experimental vaccines have used the Josiah strain of LASV as platform and only a few have been assessed against a nonhomologous LASV challenge [115]. It is possible that both cross-reactive T and B cell epitopes are needed for broad protection.
There has been little MCM development for LUJV, LCMV, and other NW arenaviruses.
It is not known whether existing arenavirus vaccines can be improved. JUNV Candid#1 is approved in endemic areas, but evidence of reversion or neurovirulence exists. The durability of protection for existing and experimental vaccines has not been determined.
Several promising recombinant live-attenuated vaccines are under evaluation in humans, but more benign platforms (eg, VLP, subunit, inactivated vaccines) may be required for immune-compromised populations.
PROTOTYPE PATHOGENS
Viruses are organized into families based on shared properties. In-depth knowledge of prototype viral pathogens within each family can inform development of MCM for other family members. The Arenaviridae family is genetically diverse, and it is unlikely that a single arenavirus prototype will be sufficient for this purpose. A representative from both OW and NW mammarenaviruses will likely be necessary. LASV is the ideal OW mammarenavirus to serve as a prototype pathogen from a public health standpoint due to its high incidence of disease, but careful consideration of phenotypic differences across phylogenetic lineages must be taken into account. Furthermore, high seroprevalence rates suggest asymptomatic or avirulent LASV (or closely related OW arenaviruses) are circulating. For MCM development, a highly virulent isolate may be preferred; however, there is value in understanding the protection conferred by “avirulent” strains. JUNV could be the ideal NW arenavirus for similar issues related to incidence; however, there have been reports of isolate-specific outcomes where some are more neurotropic, others more viscerotropic, and others a mix of both.
Knowledge Gaps
It is unclear which isolates of LASV and JUNV are most representative of contemporary natural infections or whether a single prototype is appropriate for either virus.
Most “prototype” isolates of LASV and JUNV were isolated between 30 and 50 years ago. It will be important to understand how much currently circulating isolates have changed and how the differences will impact MCM efficacy. A challenge will be to access recent isolates, which is heavily dependent on active surveillance systems with cooperative international collaboration.
While there is an appropriate focus on the most pathogenic members of the Arenaviridae family, treatment options for other arenaviruses, except JUNV, remain limited. For example, immunocompromised patients infected with LCMV do not have access to specific antivirals.
CONCLUSIONS
Improved, though still limited, clinical and laboratory research infrastructure in the Lassa endemic zone of West Africa has created a pathway to development of effective countermeasures, including small-molecule drugs, immunotherapeutics, and vaccines for LASV. Both LASV vaccines and drugs or drug combinations are urgently needed. Development for MCM for other OW arenaviruses, in particular LCMV, should also be a priority. Moreover, while development of MCM for JUNV preceded that for LASV by decades, development of countermeasures against other NW arenaviruses still represents an important gap. Designation of LASV and JUNV as prototype pathogens can facilitate progress in addressing public health challenges posed by members of the Arenaviridae family.
Notes
Author contributions. All authors wrote the original draft, and reviewed and edited the article.
Acknowledgments. The authors gratefully acknowledge the Viral Hemorrhagic Fever Consortium, the Viral Hemorrhagic Fever Immunotherapeutic Consortium, and patients and families affected by arenavirus infections.
Financial support. This work was supported by National Institutes of Health (NIH) (grant numbers R21 AI137809 to K. M. H.; U01AI51801 to R. W. C.; U19 A142790, R01AI132244, R01AI141251, and U19 AI142790 to R. W. C., E. O. S., and R. F. G.; U19AI135995, U01AI151812, and UL1TR002550 to K. G. A.; U54CA260581, U54HG007480, OT2HL158260, U19AI135995, and U01AI151812 to R. F. G.); and the Coalition for Epidemic Preparedness Innovation, the Wellcome Trust Foundation, Gilead Sciences, and the European and Developing Countries Clinical Trials Partnership Programme (to R. F. G).
Supplement sponsorship . This article appears as part of the supplement “Pandemic Preparedness at NIAID: Prototype Pathogen Approach to Accelerate Medical Countermeasures—Vaccines and Monoclonal Antibodies,” sponsored by the National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD.
Contributor Information
Kathryn M Hastie, Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA.
Lilia I Melnik, Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA.
Robert W Cross, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston National Laboratory, Galveston, Texas, USA.
Raphaëlle M Klitting, Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA; Scripps Research Translational Institute, La Jolla, California, USA.
Kristian G Andersen, Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA; Scripps Research Translational Institute, La Jolla, California, USA.
Erica Ollmann Saphire, Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA; Department of Medicine, University of California San Diego, La Jolla, California, USA.
Robert F Garry, Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Zalgen Labs LLC, Frederick, Maryland, USA.
References
- 1. Radoshitzky SR, Bào Y, Buchmeier MJ, et al. Past, present, and future of arenavirus taxonomy. Arch Virol 2015; 160:1851–74. doi: 10.1007/s00705-015-2418-y [DOI] [PubMed] [Google Scholar]
- 2. Loayza Mafayle R, Morales-Betoulle ME, Romero C, et al. Chapare hemorrhagic fever and virus detection in rodents in Bolivia in 2019. N Engl J Med 2022; 386:2283–94. doi: 10.1056/NEJMoa2110339 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Cajimat MN, Milazzo ML, Mauldin MR, Bradley RD, Fulhorst CF. Diversity among Tacaribe serocomplex viruses (family Arenaviridae) associated with the southern plains woodrat (Neotoma micropus). Virus Res 2013; 178:486–94. doi: 10.1016/j.virusres.2013.10.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Bowen MD, Peters CJ, Nichol ST. Phylogenetic analysis of the Arenaviridae: patterns of virus evolution and evidence for cospeciation between arenaviruses and their rodent hosts. Mol Phylogenet Evol 1997; 8:301–16. doi: 10.1006/mpev.1997.0436 [DOI] [PubMed] [Google Scholar]
- 5. Kronmann KC, Nimo-Paintsil S, Guirguis F, et al. Two novel arenaviruses detected in pygmy mice, Ghana. Emerg Infect Dis 2013; 19:1832–5. doi: 10.3201/eid1911.121491 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Olayemi A, Cadar D, Magassouba N, et al. New hosts of the Lassa virus. Sci Rep 2016; 6:25280. doi: 10.1038/srep25280 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Monath TP, Newhouse VF, Kemp GE, Setzer HW, Cacciapuoti A. Lassa virus isolation from Mastomys natalensis rodents during an epidemic in Sierra Leone. Science 1974; 185:263–5. doi: 10.1126/science.185.4147.263 [DOI] [PubMed] [Google Scholar]
- 8. Li K, Lin XD, Li MHet al. Genomic analysis of Wenzhou virus in rodents from Zhejiang province. Zhonghua Liu Xing Bing Xue Za Zhi 2017; 38:384–7. [in Chinese]. [DOI] [PubMed] [Google Scholar]
- 9. Blasdell KR, Becker SD, Hurst J, Begon M, Bennett M. Host range and genetic diversity of arenaviruses in rodents, United Kingdom. Emerg Infect Dis 2008; 14:1455–8. doi: 10.3201/eid1409.080209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Li K, Lin X-D, Wang W, et al. Isolation and characterization of a novel arenavirus harbored by rodents and shrews in Zhejiang province, China. Virology 2015; 476:37–42. doi: 10.1016/j.virol.2014.11.026 [DOI] [PubMed] [Google Scholar]
- 11. Ibukun FI. Inter-lineage variation of Lassa virus glycoprotein epitopes: a challenge to Lassa virus vaccine development. Viruses 2020; 12:386. doi: 10.3390/v12040386 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Charrel RN, Feldmann H, Fulhorst CF, Khelifa R, de Chesse R, de Lamballerie X. Phylogeny of new world arenaviruses based on the complete coding sequences of the small genomic segment identified an evolutionary lineage produced by intrasegmental recombination. Biochem Biophys Res Commun 2002; 296:1118–24. doi: 10.1016/S0006-291X(02)02053-3 [DOI] [PubMed] [Google Scholar]
- 13. Downs WG, Anderson CR, Spence L, Aitken TH, Greenhall AH. Tacaribe virus, a new agent isolated from Artibeus bats and mosquitoes in Trinidad, West Indies. Am J Trop Med Hyg 1963; 12:640–6. doi: 10.4269/ajtmh.1963.12.640 [DOI] [PubMed] [Google Scholar]
- 14. Brisse ME, Ly H. Hemorrhagic fever-causing arenaviruses: lethal pathogens and potent immune suppressors. Front Immunol 2019; 10:372. doi: 10.3389/fimmu.2019.00372 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Milazzo ML, Cajimat MN, Mauldin MR, et al. Epizootiology of Tacaribe serocomplex viruses (Arenaviridae) associated with neotomine rodents (Cricetidae, Neotominae) in southern California. Vector Borne Zoonotic Dis 2015; 15:156–66. doi: 10.1089/vbz.2014.1625 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Fulhorst CF, Bowen MD, Salas RA, et al. Natural rodent host associations of Guanarito and pirital viruses (family Arenaviridae) in central Venezuela. Am J Trop Med Hyg 1999; 61:325–30. doi: 10.4269/ajtmh.1999.61.325 [DOI] [PubMed] [Google Scholar]
- 17. Maes P, Adkins S, Alkhovsky SV, et al. Taxonomy of the order Bunyavirales: second update 2018. Arch Virol 2019; 164:927–41. doi: 10.1007/s00705-018-04127-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Maes P, Alkhovsky SV, Bào Y, et al. Taxonomy of the family Arenaviridae and the order Bunyavirales: update 2018. Arch Virol 2018; 163:2295–310. doi: 10.1007/s00705-018-3843-5 [DOI] [PubMed] [Google Scholar]
- 19. Shi M, Lin XD, Chen X, et al. The evolutionary history of vertebrate RNA viruses. Nature 2018; 556:197–202. doi: 10.1038/s41586-018-0012-7 [DOI] [PubMed] [Google Scholar]
- 20. Mordecai GJ, Miller KM, Di Cicco E, et al. Endangered wild salmon infected by newly discovered viruses. Elife 2019; 8:e47615. doi: 10.7554/eLife.47615 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Abudurexiti A, Adkins S, Alioto D, et al. Taxonomy of the order Bunyavirales: update 2019. Arch Virol 2019; 164:1949–65. doi: 10.1007/s00705-019-04253-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Pontremoli C, Forni D, Sironi M. Arenavirus genomics: novel insights into viral diversity, origin, and evolution. Curr Opin Virol 2019; 34:18–28. doi: 10.1016/j.coviro.2018.11.001 [DOI] [PubMed] [Google Scholar]
- 23. Hepojoki J, Salmenperä P, Sironen T, et al. Arenavirus coinfections are common in snakes with boid inclusion body disease. J Virol 2015; 89:8657–60. doi: 10.1128/JVI.01112-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Hastie KM, Zandonatti MA, Kleinfelter LM, et al. Structural basis for antibody-mediated neutralization of Lassa virus. Science 2017; 356:923–8. doi: 10.1126/science.aam7260 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Vogel D, Rosenthal M, Gogrefe N, Reindl S, Günther S. Biochemical characterization of the Lassa virus L protein. J Biol Chem 2019; 294:8088–100. doi: 10.1074/jbc.RA118.006973 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Robinson JE, Hastie KM, Cross RW, et al. Most neutralizing human monoclonal antibodies target novel epitopes requiring both Lassa virus glycoprotein subunits. Nat Commun 2016; 7:11544. doi: 10.1038/ncomms11544 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Stenglein MD, Sanders C, Kistler AL, et al. Identification, characterization, and in vitro culture of highly divergent arenaviruses from boa constrictors and annulated tree boas: candidate etiological agents for snake inclusion body disease. mBio 2012; 3:e00180-12. doi: 10.1128/mBio.00180-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Hastie KM, Liu T, Li S, et al. Crystal structure of the Lassa virus nucleoprotein-RNA complex reveals a gating mechanism for RNA binding. Proc Natl Acad Sci U S A 2011; 108:19365–70. doi: 10.1073/pnas.1108515108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Hastie KM, Kimberlin CR, Zandonatti MA, MacRae IJ, Saphire EO. Structure of the Lassa virus nucleoprotein reveals a dsRNA-specific 3′ to 5′ exonuclease activity essential for immune suppression. Proc Natl Acad Sci U S A 2011; 108:2396–401. doi: 10.1073/pnas.1016404108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Reynard S, Russier M, Fizet A, Carnec X, Baize S. Exonuclease domain of the Lassa virus nucleoprotein is critical to avoid RIG-I signaling and to inhibit the innate immune response. J Virol 2014; 88:13923–7. doi: 10.1128/JVI.01923-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Qi X, Lan S, Wang W, et al. Cap binding and immune evasion revealed by Lassa nucleoprotein structure. Nature 2010; 468:779–83. doi: 10.1038/nature09605 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Hastie KM, King LB, Zandonatti MA, Saphire EO. Structural basis for the dsRNA specificity of the Lassa virus NP exonuclease. PLoS One 2012; 7:e44211. doi: 10.1371/journal.pone.0044211 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Hastie KM, Zandonatti M, Liu T, Li S, Woods VL Jr, Saphire EO. Crystal structure of the oligomeric form of Lassa virus matrix protein Z. J Virol 2016; 90:4556–62. doi: 10.1128/JVI.02896-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Volpon L, Osborne MJ, Capul AA, de la Torre JC, Borden KL. Structural characterization of the Z RING-eIF4E complex reveals a distinct mode of control for eIF4E. Proc Natl Acad Sci U S A 2010; 107:5441–6. doi: 10.1073/pnas.0909877107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Garry CE, Garry RF. Proteomics computational analyses suggest that the antennavirus glycoprotein complex includes a class I viral fusion protein (α-penetrene) with an internal zinc-binding domain and a stable signal peptide. Viruses 2019; 11:750. doi: 10.3390/v11080750 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Kang H, Cong J, Wang C, et al. Structural basis for recognition and regulation of arenavirus polymerase L by Z protein. Nat Commun 2021; 12:4134. doi: 10.1038/s41467-021-24458-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Ma J, Zhang S, Zhang X. Structure of Machupo virus polymerase in complex with matrix protein Z. Nat Commun 2021; 12:6163. doi: 10.1038/s41467-021-26432-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Xu X, Peng R, Peng Q, et al. Cryo-EM structures of Lassa and Machupo virus polymerases complexed with cognate regulatory Z proteins identify targets for antivirals. Nat Microbiol 2021; 6:921–31. doi: 10.1038/s41564-021-00916-w [DOI] [PubMed] [Google Scholar]
- 39. Kouba T, Vogel D, Thorkelsson SR, et al. Conformational changes in Lassa virus L protein associated with promoter binding and RNA synthesis activity. Nat Commun 2021; 12:7018. doi: 10.1038/s41467-021-27305-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Joseph S, Campbell KP. Lassa fever virus binds matriglycan-A polymer of alternating xylose and glucuronate-on α-dystroglycan. Viruses 2021; 13:1679. doi: 10.3390/v13091679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Brouillette RB, Phillips EK, Patel R, et al. TIM-1 mediates dystroglycan-independent entry of Lassa virus. J Virol 2018; 92:e00093-18. doi: 10.1128/JVI.00093-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Fedeli C, Moreno H, Kunz S. The role of receptor tyrosine kinases in Lassa virus cell entry. Viruses 2020; 12:857. doi: 10.3390/v12080857 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Fedeli C, Torriani G, Galan-Navarro C, et al. Axl can serve as entry factor for Lassa virus depending on the functional glycosylation of dystroglycan. J Virol 2018; 92:e01613-1. doi: 10.1128/JVI.01613-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Goncalves AR, Moraz ML, Pasquato A, Helenius A, Lozach PY, Kunz S. Role of DC-SIGN in Lassa virus entry into human dendritic cells. J Virol 2013; 87:11504–15. doi: 10.1128/JVI.01893-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Cao W, Henry MD, Borrow P, et al. Identification of alpha-dystroglycan as a receptor for lymphocytic choriomeningitis virus and Lassa fever virus. Science 1998; 282:2079–81. doi: 10.1126/science.282.5396.2079 [DOI] [PubMed] [Google Scholar]
- 46. Li S, Sun Z, Pryce R, et al. Acidic pH-induced conformations and LAMP1 binding of the Lassa virus glycoprotein spike. PLoS Pathog 2016; 12:e1005418. doi: 10.1371/journal.ppat.1005418 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Israeli H, Cohen-Dvashi H, Shulman A, Shimon A, Diskin R. Mapping of the Lassa virus LAMP1 binding site reveals unique determinants not shared by other old world arenaviruses. PLoS Pathog 2017; 13:e1006337. doi: 10.1371/journal.ppat.1006337 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Jae LT, Raaben M, Herbert AS, et al. Virus entry. Lassa virus entry requires a trigger-induced receptor switch. Science 2014; 344:1506–10. doi: 10.1126/science.1252480 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Hulseberg CE, Fénéant L, Szymańska KM, White JM. Lamp1 increases the efficiency of Lassa virus infection by promoting fusion in less acidic endosomal compartments. mBio 2018; 9:e01818-17. doi: 10.1128/mBio.01818-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Bakkers MJG, Moon-Walker A, Herlo R, et al. CD164 is a host factor for lymphocytic choriomeningitis virus entry. Proc Natl Acad Sci U S A 2022; 119:e2119676119. doi: 10.1073/pnas.2119676119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Raaben M, Jae LT, Herbert AS, et al. NRP2 and CD63 are host factors for Lujo virus cell entry. Cell Host Microbe 2017; 22:688–96.e5. doi: 10.1016/j.chom.2017.10.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Reguera J, Gerlach P, Rosenthal M, et al. Comparative structural and functional analysis of Bunyavirus and Arenavirus cap-snatching endonucleases. PLoS Pathog 2016; 12:e1005636. doi: 10.1371/journal.ppat.1005636 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Ogbu O, Ajuluchukwu E, Uneke CJ. Lassa fever in West African sub-region: an overview. J Vector Borne Dis 2007; 44:1–11. [PubMed] [Google Scholar]
- 54. Richmond JK, Baglole DJ. Lassa fever: epidemiology, clinical features, and social consequences. BMJ 2003; 327:1271–5. doi: 10.1136/bmj.327.7426.1271 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Macher AM, Wolfe MS. Historical Lassa fever reports and 30-year clinical update. Emerg Infect Dis 2006; 12:835–7. doi: 10.3201/eid1205.050052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Parodi AS, Greenway DY, Rugiero HR, et al. Una nueva enfermedad epidémica a germen desconocido: hipertermia nefrotóxica, leucopénica, y enantemática. Dia Médico 1955; 27:1204–10. [PubMed] [Google Scholar]
- 57. Mills JN, Childs JE. Ecologic studies of rodent reservoirs: their relevance for human health. Emerg Infect Dis 1998; 4:529–37. doi: 10.3201/eid0404.980403 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Porcasi X, Calderón GE, Lamfri M, Scavuzzo M, Sabattini MS, Polop JJ. Predictive distribution maps of rodent reservoir species of zoonoses in southern America. Mastozoología Neotropical 2005; 12:199–216. [Google Scholar]
- 59. Briggiler A, Sinchi A, Coronel F, et al. New transmission scenarios of the Argentine hemorrhagic fever since the introduction of the live attenuated Junin virus vaccine (Candid #1): an experience in migrant workers. Rev Peru Med Exp Salud Publica 2015; 32:165–71. [in Spanish]. doi: 10.17843/rpmesp.2015.321.1589 [DOI] [PubMed] [Google Scholar]
- 60. Piacenza MF, Calderón GE, Enría Det al. Spatial difference in the incidence of Argentine hemorrhagic fever and composition and abundance of rodents in the assemblage. Rev Chilena Infectol 2018; 35:386–94. [in Spanish]. doi: 10.4067/s0716-10182018000400386 [DOI] [PubMed] [Google Scholar]
- 61. Frank MG, Beitscher A, Webb CM, Raabe V. South American hemorrhagic fevers: a summary for clinicians. Int J Infect Dis 2021; 105:505–15. doi: 10.1016/j.ijid.2021.02.046 [DOI] [PubMed] [Google Scholar]
- 62. Knobloch J, McCormick JB, Webb PA, Dietrich M, Schumacher HH, Dennis E. Clinical observations in 42 patients with Lassa fever. Tropenmed Parasitol 1980; 31:389–98. [PubMed] [Google Scholar]
- 63. McCormick JB, Fisher-Hoch SP. Lassa fever. Curr Top Microbiol Immunol 2002; 262:75–109. [DOI] [PubMed] [Google Scholar]
- 64. Ilori EA, Frank C, Dan-Nwafor CC, et al. Increase in Lassa fever cases in Nigeria, January-March 2018. Emerg Infect Dis 2019; 25:1026–7. doi: 10.3201/eid2505.181247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Okokhere P, Colubri A, Azubike C, et al. Clinical and laboratory predictors of Lassa fever outcome in a dedicated treatment facility in Nigeria: a retrospective, observational cohort study. Lancet Infect Dis 2018; 18:684–95. doi: 10.1016/S1473-3099(18)30121-X [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Abba Y, Hassim H, Hamzah H, et al. In vitro isolation and molecular identification of reptarenavirus in Malaysia. Virus Genes 2016; 52:640–50. doi: 10.1007/s11262-016-1345-7 [DOI] [PubMed] [Google Scholar]
- 67. Keller S, Hetzel U, Sironen T, et al. Co-infecting reptarenaviruses can be vertically transmitted in Boa constrictor. PLoS Pathog 2017; 13:e1006179. doi: 10.1371/journal.ppat.1006179 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Bodewes R, Raj VS, Kik MJ, et al. Updated phylogenetic analysis of arenaviruses detected in boid snakes. J Virol 2014; 88:1399–400. doi: 10.1128/JVI.02753-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Hetzel U, Sironen T, Laurinmäki P, et al. Isolation, identification, and characterization of novel arenaviruses, the etiological agents of boid inclusion body disease. J Virol 2013; 87:10918–35. doi: 10.1128/JVI.01123-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Hetzel U, Sironen T, Laurinmäki P, et al. Reply to “Updated phylogenetic analysis of arenaviruses detected in boid snakes”. J Virol 2014; 88:1401. doi: 10.1128/JVI.03044-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Hepojoki J, Hepojoki S, Smura T, et al. Characterization of Haartman Institute snake virus-1 (HISV-1) and HISV-like viruses—the representatives of genus Hartmanivirus, family Arenaviridae. PLoS Pathog 2018; 14:e1007415. doi: 10.1371/journal.ppat.1007415 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Watanabe Y, Raghwani J, Allen JD, et al. Structure of the Lassa virus glycan shield provides a model for immunological resistance. Proc Natl Acad Sci U S A 2018; 115:7320–5. doi: 10.1073/pnas.1803990115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Lenz O, ter Meulen J, Klenk HD, Seidah NG, Garten W. The Lassa virus glycoprotein precursor GP-C is proteolytically processed by subtilase SKI-1/S1P. Proc Natl Acad Sci U S A 2001; 98:12701–5. doi: 10.1073/pnas.221447598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Tang-Huau TL, Feldmann H, Rosenke K. Animal models for Lassa virus infection. Curr Opin Virol 2019; 37:112–7. doi: 10.1016/j.coviro.2019.07.005 [DOI] [PubMed] [Google Scholar]
- 75. Sattler RA, Paessler S, Ly H, Huang C. Animal models of Lassa fever. Pathogens 2020; 9:197. doi: 10.3390/pathogens9030197 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Walker DH, Wulff H, Lange JV, Murphy FA. Comparative pathology of Lassa virus infection in monkeys, guinea-pigs, and Mastomys natalensis. Bull World Health Organ 1975; 52:523–34. [PMC free article] [PubMed] [Google Scholar]
- 77. Jahrling PB, Smith S, Hesse RA, Rhoderick JB. Pathogenesis of Lassa virus infection in guinea pigs. Infect Immun 1982; 37:771–8. doi: 10.1128/iai.37.2.771-778.1982 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Bell TM, Shaia CI, Bearss JJ, et al. Temporal progression of lesions in Guinea pigs infected with Lassa virus. Vet Pathol 2017; 54:549–62. doi: 10.1177/0300985816677153 [DOI] [PubMed] [Google Scholar]
- 79. Cross RW, Mire CE, Branco LM, et al. Treatment of Lassa virus infection in outbred guinea pigs with first-in-class human monoclonal antibodies. Antiviral Res 2016; 133:218–22. doi: 10.1016/j.antiviral.2016.08.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Yun NE, Ronca S, Tamura A, et al. Animal model of sensorineural hearing loss associated with Lassa virus infection. J Virol 2015; 90:2920–7. doi: 10.1128/JVI.02948-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Hickerson BT, Sefing EJ, Bailey KW, Van Wettere AJ, Penichet ML, Gowen BB. Type I interferon underlies severe disease associated with Junín virus infection in mice. Elife 2020; 9:e55352. doi: 10.7554/eLife.55352 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Hensley LE, Smith MA, Geisbert JB, et al. Pathogenesis of Lassa fever in cynomolgus macaques. Virol J 2011; 8:205. doi: 10.1186/1743-422X-8-205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Baize S, Marianneau P, Loth P, et al. Early and strong immune responses are associated with control of viral replication and recovery in Lassa virus-infected cynomolgus monkeys. J Virol 2009; 83:5890–903. doi: 10.1128/JVI.01948-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Cashman KA, Wilkinson ER, Zeng X, et al. Immune-mediated systemic vasculitis as the proposed cause of sudden-onset sensorineural hearing loss following Lassa virus exposure in cynomolgus macaques. mBio 2018; 9:e01896-18. doi: 10.1128/mBio.01896-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Zhang S. America is running low on a crucial resource for COVID-19 vaccines. The Atlantic, August 21, 2020. [Google Scholar]
- 86. Boisen ML, Uyigue E, Aiyepada J, et al. Field evaluation of a Pan-Lassa rapid diagnostic test during the 2018 Nigerian Lassa fever outbreak. Sci Rep 2020; 10:8724. doi: 10.1038/s41598-020-65736-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Branco LM, Grove JN, Boisen ML, et al. Emerging trends in Lassa fever: redefining the role of immunoglobulin M and inflammation in diagnosing acute infection. Virol J 2011; 8:478. doi: 10.1186/1743-422X-8-478 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Enria DA, Briggiler AM, Sánchez Z. Treatment of Argentine hemorrhagic fever. Antiviral Res 2008; 78:132–9. doi: 10.1016/j.antiviral.2007.10.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Enría D, Franco SG, Ambrosio A, Vallejos D, Levis S, Maiztegui J. Current status of the treatment of Argentine hemorrhagic fever. Med Microbiol Immunol 1986; 175:173–6. doi: 10.1007/BF02122443 [DOI] [PubMed] [Google Scholar]
- 90. Jahrling PB, Peters CJ, Stephen EL. Enhanced treatment of Lassa fever by immune plasma combined with ribavirin in cynomolgus monkeys. J Infect Dis 1984; 149:420–7. doi: 10.1093/infdis/149.3.420 [DOI] [PubMed] [Google Scholar]
- 91. Jahrling PB, Peters CJ. Passive antibody therapy of Lassa fever in cynomolgus monkeys: importance of neutralizing antibody and Lassa virus strain. Infect Immun 1984; 44:528–33. doi: 10.1128/iai.44.2.528-533.1984 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. McCormick JB, King IJ, Webb PA, et al. Lassa fever. Effective therapy with ribavirin. N Engl J Med 1986; 314:20–6. doi: 10.1056/NEJM198601023140104 [DOI] [PubMed] [Google Scholar]
- 93. Mire CE, Cross RW, Geisbert JB, et al. Human-monoclonal-antibody therapy protects nonhuman primates against advanced Lassa fever. Nat Med 2017; 23:1146–9. doi: 10.1038/nm.4396 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Cross RW, Hastie KM, Mire CE, et al. Antibody therapy for Lassa fever. Curr Opin Virol 2019; 37:97–104. doi: 10.1016/j.coviro.2019.07.003 [DOI] [PubMed] [Google Scholar]
- 95. Borenstein-Katz A, Shulman A, Hamawi H, Leitner O, Diskin R. Differential antibody-based immune response against isolated GP1 receptor-binding domains from Lassa and Junín viruses. J Virol 2019; 93:e00090-19. doi: 10.1128/JVI.00090-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Cresta B, Padula P, de Martinez Segovia M. Biological properties of Junin virus proteins. I. Identification of the immunogenic glycoprotein. Intervirology 1980; 13:284–8. doi: 10.1159/000149136 [DOI] [PubMed] [Google Scholar]
- 97. Sanchez A, Pifat DY, Kenyon RH, Peters CJ, McCormick JB, Kiley MP. Junin virus monoclonal antibodies: characterization and cross-reactivity with other arenaviruses. J Gen Virol 1989; 70:1125–32. doi: 10.1099/0022-1317-70-5-1125 [DOI] [PubMed] [Google Scholar]
- 98. Clark LE, Mahmutovic S, Raymond DD, et al. Vaccine-elicited receptor-binding site antibodies neutralize two new world hemorrhagic fever arenaviruses. Nat Commun 2018; 9:1884. doi: 10.1038/s41467-018-04271-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Zeitlin L, Cross RW, Geisbert JB, et al. Therapy for Argentine hemorrhagic fever in nonhuman primates with a humanized monoclonal antibody. Proc Natl Acad Sci U S A 2021; 118:e2023332118. doi: 10.1073/pnas.2023332118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Amanat F, Duehr J, Huang C, Paessler S, Tan GS, Krammer F. Monoclonal antibodies with neutralizing activity and Fc-effector functions against the Machupo virus glycoprotein. J Virol 2020; 94:e01741-19. doi: 10.1128/JVI.01741-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Enria DA, Ambrosio AM, Briggiler AM, et al. , Study Group on Argentine Hemorrhagic Fever Vaccine . Candid#1 vaccine against Argentine hemorrhagic fever produced in Argentina. Immunogenicity and safety. Medicina (B Aires) 2010; 70:215–22. [in Spanish]. [PubMed] [Google Scholar]
- 102. Gowen BB, Hickerson BT, York J, et al. Second-generation live-attenuated Candid#1 vaccine virus resists reversion and protects against lethal Junín virus infection in Guinea pigs. J Virol 2021; 95:e0039721. doi: 10.1128/JVI.00397-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Johnson DM, Cubitt B, Pfeffer TL, de la Torre JC, Lukashevich IS. Lassa virus vaccine candidate ML29 generates truncated viral RNAs which contribute to interfering activity and attenuation. Viruses 2021; 13:214. doi: 10.3390/v13020214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Cross RW, Xu R, Matassov D, et al. Quadrivalent VesiculoVax vaccine protects nonhuman primates from viral-induced hemorrhagic fever and death. J Clin Invest 2020; 130:539–51. doi: 10.1172/JCI131958 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Stein DR, Warner BM, Soule G, et al. A recombinant vesicular stomatitis-based Lassa fever vaccine elicits rapid and long-term protection from lethal Lassa virus infection in guinea pigs. NPJ Vaccines 2019; 4:8. doi: 10.1038/s41541-019-0104-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Cashman KA, Wilkinson ER, Shaia CI, et al. A DNA vaccine delivered by dermal electroporation fully protects cynomolgus macaques against Lassa fever. Hum Vaccin Immunother 2017; 13:2902–11. doi: 10.1080/21645515.2017.1356500 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Mateo M, Reynard S, Journeaux A, et al. A single-shot Lassa vaccine induces long-term immunity and protects cynomolgus monkeys against heterologous strains. Sci Transl Med 2021; 13:eabf6348. doi: 10.1126/scitranslmed.abf6348 [DOI] [PubMed] [Google Scholar]
- 108. Salvato MS, Domi A, Guzmán-Cardozo C, et al. A single dose of modified vaccinia ankara expressing Lassa virus-like particles protects mice from lethal intra-cerebral virus challenge. Pathogens 2019; 8:133. doi: 10.3390/pathogens8030133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Abreu-Mota T, Hagen KR, Cooper K, et al. Non-neutralizing antibodies elicited by recombinant Lassa-rabies vaccine are critical for protection against Lassa fever. Nat Commun 2018; 9:4223. doi: 10.1038/s41467-018-06741-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Wang M, Li R, Li Y, et al. Construction and immunological evaluation of an adenoviral vector-based vaccine candidate for Lassa fever. Viruses 2021; 13:484. doi: 10.3390/v13030484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Fischer RJ, Purushotham JN, van Doremalen N, et al. ChAdOx1-vectored Lassa fever vaccine elicits a robust cellular and humoral immune response and protects guinea pigs against lethal Lassa virus challenge. NPJ Vaccines 2021; 6:32. doi: 10.1038/s41541-021-00291-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Müller H, Fehling SK, Dorna J, et al. Adjuvant formulated virus-like particles expressing native-like forms of the Lassa virus envelope surface glycoprotein are immunogenic and induce antibodies with broadly neutralizing activity. NPJ Vaccines 2020; 5:71. doi: 10.1038/s41541-020-00219-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Branco LM, Grove JN, Geske FJ, et al. Lassa virus-like particles displaying all major immunological determinants as a vaccine candidate for Lassa hemorrhagic fever. Virol J 2010; 7:279. doi: 10.1186/1743-422X-7-279 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Spengler JR, Kainulainen MH, Welch SR, et al. Lassa virus replicon particle vaccine protects strain 13/N guinea pigs against challenge with geographically and genetically diverse viral strains. J Infect Dis 2023; 226:1545–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Olschläger S, Flatz L. Vaccination strategies against highly pathogenic arenaviruses: the next steps toward clinical trials. PLoS Pathog 2013; 9:e1003212. doi: 10.1371/journal.ppat.1003212 [DOI] [PMC free article] [PubMed] [Google Scholar]