ABSTRACT
Several highly pathogenic mammarenaviruses cause severe hemorrhagic and neurologic disease in humans for which vaccines and antivirals are limited or unavailable. New World (NW) mammarenavirus Machupo virus (MACV) infection causes Bolivian hemorrhagic fever in humans. We previously reported that the disruption of specific N-linked glycan sites on the glycoprotein (GPC) partially attenuates MACV in an interferon alpha/beta and gamma (IFN-α/ß and -γ) receptor knockout (R-/-) mouse model. However, some capability to induce neurological pathology still remained. The highly pathogenic Junin virus (JUNV) is another NW arenavirus closely related to MACV. An F427I substitution in the GPC transmembrane domain (TMD) rendered JUNV attenuated in a lethal mouse model after intracranial inoculation. In this study, we rationally designed and rescued a MACV containing mutations at two glycosylation sites and the corresponding F438I substitution in the GPC TMD. The MACV mutant is fully attenuated in IFN-α/ß and -γ R-/- mice and outbred guinea pigs. Furthermore, inoculation with this mutant MACV completely protected guinea pigs from wild-type MACV lethal challenge. Last, we found the GPC TMD F438I substitution greatly impaired MACV growth in neuronal cell lines of mouse and human origins. Our results highlight the critical roles of the glycans and the TMD on the GPC in arenavirus virulence, which provide insight into the rational design of potential vaccine candidates for highly pathogenic arenaviruses. IMPORTANCE For arenaviruses, the only vaccine available is the live attenuated Candid#1 vaccine, a JUNV vaccine approved in Argentina. We and others have found that the glycans on GPC and the F427 residue in the GPC TMD are important for virulence of JUNV. Nevertheless, mutating either of them is not sufficient for full and stable attenuation of JUNV. Using reverse genetics, we disrupted specific glycosylation sites on MACV GPC and also introduced the corresponding F438I substitution in the GPC TMD. This MACV mutant is fully attenuated in two animal models and protects animals from lethal infection. Thus, our studies highlight the feasibility of rational attenuation of highly pathogenic arenaviruses for vaccine development. Another important finding from this study is that the F438I substitution in GPC TMD could substantially affect MACV replication in neurons. Future studies are warranted to elucidate the underlying mechanism and the implication of this mutation in arenavirus neural tropism.
Subject(s)
Arenaviruses, New World , Hemorrhagic Fever, American , Viral Vaccines , Animals , Arenaviruses, New World/genetics , Arenaviruses, New World/immunology , Disease Models, Animal , Glycoproteins/metabolism , Glycosylation , Guinea Pigs , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/virology , Junin virus/genetics , Junin virus/immunology , Mutation , Vaccines, Attenuated/immunology , Viral Vaccines/immunologyABSTRACT
Candid#1 is the first live attenuated vaccine produced and registered in Argentina. Produced since 2003 at the INEVH to prevent Argentine hemorrhagic fever, it is obtained by harvesting supernatants of diploid cells infected with an attenuated strain of Junin virus and subsequent lyophilization. The stability of this vaccine is crucial to ensure its effectiveness. This study was aimed to evaluate the stability of Candid#1 by exposing it to different time and temperature conditions. Three vaccine batches produced in 2003 were analysed according to the following storage scheme: (a) reconstituted vaccine at 2 °C to 8 °C for 8 days; (b) lyophilized vaccine at 2 °C to 8 °C for 6 months; (c) lyophilized vaccine at -18 °C to -20 °C for 10 years. The potency was assessed in Vero cell monolayers under agar. The results were: (a) reconstituted vaccine was stable between 2 °C and 8 °C for 8 days, (b) lyophilized vaccine was stable between 2 °C and 8 °C for 2 months, and (c) lyophilized vaccine was stable 9 years between -18 °C and -20 °C, keeping all its properties. These results allowed us to establish the following storage conditions and expiration times for Candid#1: (a) reconstituted: 12 hours between 2 °C and 8 °C, (b) lyophilized: 30 days between 2 °C and 8 °C and (c) lyophilized: 9 years between -18 °C and -20 °C. Based on our results, favorable changes were made in the conditions of transport, storage and distribution of the vaccine. Domestic freezers in strategically located centers were installed, allowing the preservation of vaccine stocks for distribution to secondary vaccination centers.
Subject(s)
Antibodies, Viral/immunology , Arenaviruses, New World/immunology , Drug Storage/methods , Hemorrhagic Fever, American/prevention & control , Viral Vaccines/immunology , Argentina , Drug Stability , Humans , Vaccines, Attenuated/immunologyABSTRACT
Candid#1 es la primera vacuna a virus vivo atenuado producida y registrada en Argentina. Se produce en el INEVH desde 2003 para prevenir la fiebre hemorrágica argentina y se obtiene mediante cosecha de sobrenadantes de cultivos de células diploides infectadas con una cepa atenuada del virus Junín, formulación y posterior liofilización. Su estabilidad es crucial para asegurar su efectividad. El objetivo de este trabajo fue evaluar la estabilidad de Candid#1 exponiéndola a distintas condiciones de temperatura y tiempo. Tres lotes producidos en 2003 fueron sometidos al siguiente esquema de almacenamiento: (a) vacuna reconstituida conservada entre 2 °C y 8 °C durante 8 días, (b) vacuna liofilizada conservada entre 2 °C y 8 °C durante 6 meses, y (c) vacuna liofilizada conservada entre -18 °C y -20 °C durante 10 años. La potencia fue evaluada en monocapa de células Vero bajo agar. Los resultados fueron: (a) Candid#1 reconstituida fue estable 8 días entre 2 °C y 8 °C, (b) Candid#1 liofilizada fue estable 2 meses entre 2 °C y 8 °C y (c) Candid#1 liofilizada fue estable 9 años entre -18 °C y -20 °C manteniendo todos sus atributos. Estos resultados permitieron establecer las siguientes condiciones de almacenamiento: reconstituida 12 horas entre 2 °C y 8 °C, liofilizada 30 días entre 2 °C y 8 °C y 9 años entre -18 °C y -20 °C. A la luz de estos resultados, se generaron cambios favorables en las condiciones de transporte, almacenamiento y distribución de la vacuna. Se implementó la instalación de freezers domésticos en centros estratégicamente distribuidos, permitiendo preservar stocks de vacuna y distribuir las dosis necesarias a vacunatorios.
Candid#1 is the first live attenuated vaccine produced and registered in Argentina. Produced since 2003 at the INEVH to prevent Argentine hemorrhagic fever, it is obtained by harvesting supernatants of diploid cells infected with an attenuated strain of Junin virus and subsequent lyophilization. The stability of this vaccine is crucial to ensure its effectiveness. This study was aimed to evaluate the stability of Candid#1 by exposing it to different time and temperature conditions. Three vaccine batches produced in 2003 were analysed according to the following storage scheme: (a) reconstituted vaccine at 2 °C to 8°C for 8 days; (b) lyophilized vaccine at 2 °C to 8 °C for 6 months; (c) lyophilized vaccine at -18 °C to -20 °C for 10 years. The potency was assessed in Vero cell monolayers under agar. The results were: (a) reconstituted vaccine was stable between 2 °C and 8 °C for 8 days, (b) lyophilized vaccine was stable between 2 °C and 8 °C for 2 months, and (c) lyophilized vaccine was stable 9 years between -18 °C and -20 °C, keeping all its properties. These results allowed us to establish the following storage conditions and expiration times for Candid#1: (a) reconstituted: 12 hours between 2 °C and 8 °C, (b) lyophilized: 30 days between 2 °C and 8 °C and (c) lyophilized: 9 years between -18 °C and -20 °C. Based on our results, favorable changes were made in the conditions of transport, storage and distribution of the vaccine. Domestic freezers in strategically located centers were installed, allowing the preservation of vaccine stocks for distribution to secondary vaccination centers.
Subject(s)
Humans , Viral Vaccines/immunology , Arenaviruses, New World/immunology , Drug Storage/methods , Hemorrhagic Fever, American/prevention & control , Antibodies, Viral/immunology , Argentina , Vaccines, Attenuated/immunology , Drug StabilityABSTRACT
Junin virus (JUNV), a highly pathogenic New World arenavirus, is the causative agent of Argentine hemorrhagic fever (AHF). The live-attenuated Candid #1 (Can) strain currently serves as a vaccine for at-risk populations. We have previously shown that the Can glycoprotein (GPC) gene is the primary gene responsible for attenuation in a guinea pig model of AHF. However, the mechanisms through which the GPC contributes to the attenuation of the Can strain remain unknown. A more complete understanding of the mechanisms underlying the attenuation and immunogenicity of the Can strain will potentially allow for the rational design of additional safe and novel vaccines. Here, we provide a detailed comparison of both RNA and protein expression profiles between both inter- and intra-segment chimeric JUNV recombinant clones expressing combinations of genes from the Can strain and the pathogenic Romero (Rom) strain. The recombinant viruses that express Can GPC, which were shown to be attenuated in guinea pigs, displayed different RNA levels and GPC processing patterns as determined by Northern and Western blot analyses, respectively. Analysis of recombinant viruses containing amino acid substitutions selected at different mouse brain passages during the generation of Can revealed that altered Can GPC processing was primarily due to the T168A substitution within G1, which eliminates an N-linked glycosylation motif. Incorporation of the T168A substitution in the Rom GPC resulted in a Can-like processing pattern of Rom GPC. In addition, JUNV GPCs containing T168A substitution were retained within the endoplasmic reticulum (ER) and displayed significantly lower cell surface expression than wild-type Rom GPC. Interestingly, the reversion A168T in Can GPC significantly increased GPC expression at the cell surface. Our results demonstrate that recombinant JUNV (rJUNV) expressing Can GPC display markedly different protein expression and elevated genomic RNA expression when compared to viruses expressing Rom GPC. Additionally, our findings indicate that the N-linked glycosylation motif at amino acid positions 166-168 is important for trafficking of JUNV GPC to the cell surface, and the elimination of this motif interferes with the GPC release from the ER.
Subject(s)
Amino Acid Motifs , Arenaviruses, New World/immunology , Glycoproteins/genetics , Glycoproteins/metabolism , Hemorrhagic Fever, American , Viral Vaccines , Animals , Arenaviruses, New World/genetics , Cell Line , Cells, Cultured , Cricetinae , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Gene Expression , Gene Expression Regulation, Viral , Glycoproteins/chemistry , Glycoproteins/immunology , Glycosylation , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/metabolism , Hemorrhagic Fever, American/prevention & control , Hemorrhagic Fever, American/virology , Humans , Protein Processing, Post-Translational , Protein Transport , Transcription, Genetic , Viral Vaccines/genetics , Viral Vaccines/immunology , VirulenceABSTRACT
UNLABELLED: Several members of the Arenaviridae can cause acute febrile diseases in humans, often resulting in lethality. The use of convalescent-phase human plasma is an effective treatment in humans infected with arenaviruses, particularly species found in South America. Despite this, little work has focused on developing potent and defined immunotherapeutics against arenaviruses. In the present study, we produced arenavirus neutralizing antibodies by DNA vaccination of rabbits with plasmids encoding the full-length glycoprotein precursors of Junín virus (JUNV), Machupo virus (MACV), and Guanarito virus (GTOV). Geometric mean neutralizing antibody titers, as measured by the 50% plaque reduction neutralization test (PRNT(50)), exceeded 5,000 against homologous viruses. Antisera against each targeted virus exhibited limited cross-species binding and, to a lesser extent, cross-neutralization. Anti-JUNV glycoprotein rabbit antiserum protected Hartley guinea pigs from lethal intraperitoneal infection with JUNV strain Romero when the antiserum was administered 2 days after challenge and provided some protection (â¼30%) when administered 4 days after challenge. Treatment starting on day 6 did not protect animals. We further formulated an IgG antibody cocktail by combining anti-JUNV, -MACV, and -GTOV antibodies produced in DNA-vaccinated rabbits. This cocktail protected 100% of guinea pigs against JUNV and GTOV lethal disease. We then expanded on this cocktail approach by simultaneously vaccinating rabbits with a combination of plasmids encoding glycoproteins from JUNV, MACV, GTOV, and Sabia virus (SABV). Sera collected from rabbits vaccinated with the combination vaccine neutralized all four targets. These findings support the concept of using a DNA vaccine approach to generate a potent pan-arenavirus immunotherapeutic. IMPORTANCE: Arenaviruses are an important family of emerging viruses. In infected humans, convalescent-phase plasma containing neutralizing antibodies can mitigate the severity of disease caused by arenaviruses, particularly species found in South America. Because of variations in potency of the human-derived product, limited availability, and safety concerns, this treatment option has essentially been abandoned. Accordingly, despite this approach being an effective postinfection treatment option, research on novel approaches to produce potent polyclonal antibody-based therapies have been deficient. Here we show that DNA-based vaccine technology can be used to make potently neutralizing antibodies in rabbits that exclusively target the glycoproteins of several human-pathogenic arenaviruses found in South America, including JUNV, MACV, GTOV, and SABV. These antibodies protected guinea pigs from lethal disease when given post-virus challenge. We also generated a purified antibody cocktail with antibodies targeting three arenaviruses and demonstrated protective efficacy against all three targets. Our findings demonstrate that use of the DNA vaccine technology could be used to produce candidate antiarenavirus neutralizing antibody-based products.
Subject(s)
Antibodies, Neutralizing/administration & dosage , Antibodies, Viral/administration & dosage , Antigens, Viral/immunology , Arenaviruses, New World/immunology , Glycoproteins/immunology , Hemorrhagic Fever, American/prevention & control , Immunization, Passive/methods , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Disease Models, Animal , Female , Guinea Pigs , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Neutralization Tests , Rabbits , Survival Analysis , Treatment Outcome , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunologyABSTRACT
Arenavirus Sabiá was originally isolated from a fatal human infection in Brazil, and after the occurrence of the second fatal human case in São Paulo state, epidemiologic and virologic studies were performed in the area where the patient lived, aiming at the identification of the Sabiá natural rodent reservoir. A broadly cross-reactive enzyme-linked immunosorbent assay (ELISA) was used to screen for antibody-positive samples. Antibodies to arenavirus were detected in two of the 55 samples of Calomys tener, and from these results, samples of rodents were analyzed by a broad RT-PCR assay. RT-PCR amplification detected arenavirus sequences in five of the 55 C. tener samples, and sequencing showed that this virus is a distinct form of Sabiá virus. Thus, we describe here the evidence for the circulation of a new arenavirus in Brazil (proposed name Pinhal virus) and its genetic characterization compared to other arenaviruses. This study also suggests C. tener as a probable rodent reservoir for this virus and associates this new virus with the lineage C of New World arenaviruses. Although we have defined some characteristics of this virus, so far, there is no evidence of its involvement in human disease.
Subject(s)
Arenaviridae Infections/veterinary , Arenaviruses, New World/isolation & purification , Sigmodontinae/virology , Animals , Antibodies, Viral/blood , Arenaviridae Infections/virology , Arenaviruses, New World/classification , Arenaviruses, New World/genetics , Arenaviruses, New World/immunology , Brazil/epidemiology , Disease Reservoirs/veterinary , Enzyme-Linked Immunosorbent Assay , PhylogenySubject(s)
Antibodies, Viral/blood , Antigens, Viral/blood , Arenaviridae Infections , Arenaviruses, New World/isolation & purification , Immunoglobulin G/blood , Rodent Diseases/epidemiology , Rodentia/virology , Animals , Arenaviridae Infections/blood , Arenaviridae Infections/epidemiology , Arenaviridae Infections/immunology , Arenaviridae Infections/veterinary , Arenaviruses, New World/immunology , Caribbean Region , Colombia , Cytochromes b/blood , Humans , Immunoassay , Rodent Diseases/blood , Rodent Diseases/immunology , Viral LoadABSTRACT
Argentine hemorrhagic fever (AHF), an acute disease caused by Junin virus (JUNV, Arenaviridae), has been an important issue to public health in Argentina since the early 1950s. The field rodent Calomys musculinus is JUNV natural reservoir and human disease is a consequence of contact with infected rodents. A steady extention of AHF endemic area is being observed since the first reports of the disease. Important achievements have been made in: (a) improvement of methods for the etiological diagnosis; (b) implementation and validation of therapeutical measures; (c) development of vaccines to protect against AHF. Reference is made to different research strategies used to obtain anti-AHF vaccines in the past and anti-arenaviral diseases in the present. Information is updated on features and field performance of Candid #1 vaccine, a live attenuted vaccine currently used to prevent AHF. This vaccine was developed through a joint international effort that envisioned it as an orphan drug. With transferred technology, Argentine government was committed to be Candid #1 manufacturer and to register this vaccine as a novel medical product under the Argentine regulatory authority. Candid #1 vaccine is the first one used to control an arenaviral hemorrhagic fever, the first live viral vaccine to be manufactured and registered in Argentina, reaching its target population through governmental effort.
Subject(s)
Arenaviruses, New World/immunology , Hemorrhagic Fever, American/prevention & control , Viral Vaccines/immunology , Animals , Argentina , Hemorrhagic Fever, American/epidemiology , Humans , Rodentia , Vaccination , Vaccines, Attenuated/immunologyABSTRACT
The evaluation of the biological and epidemiological properties of Ebola, Marburg, Lassa, and Machupo viruses suggests that they are of social importance for health care authorities. The studies have created prerequisites to the development of reliable biosafety means against these pathogens. Particular emphasis is laid on the methods for infection diagnosis and on the studies to design specific protective agents--immunoglobulins and inactivated vaccines.
Subject(s)
Arenaviruses, New World/pathogenicity , Disease Outbreaks , Ebolavirus/pathogenicity , Hemorrhagic Fever, American , Hemorrhagic Fevers, Viral , Lassa virus/pathogenicity , Marburgvirus/pathogenicity , Animals , Antibody Specificity/immunology , Antigens, Viral/administration & dosage , Antiviral Agents/therapeutic use , Arenaviruses, New World/immunology , Ebolavirus/immunology , Global Health , Hemorrhagic Fever, American/epidemiology , Hemorrhagic Fever, American/pathology , Hemorrhagic Fever, American/therapy , Hemorrhagic Fevers, Viral/epidemiology , Hemorrhagic Fevers, Viral/pathology , Hemorrhagic Fevers, Viral/therapy , Horses , Humans , Immunization , Immunoglobulins/immunology , Immunoglobulins/therapeutic use , Laboratory Infection/epidemiology , Lassa virus/immunology , Marburgvirus/immunology , Ribavirin/therapeutic use , Viral Vaccines/administration & dosage , VirulenceABSTRACT
Neutralizing antibody (NT Ab) titers to Candid #1 (C#1) vaccine against Argentine hemorrhagic fever were studied for 2 years post-vaccination in 330 volunteers, to assess whether the kinetics and/or magnitude of this immune response is modified by previous infection with the arena viruses Junin (JUN) and lymphocytic choriomeningitis (LCM). A total of 160 volunteers received C#1, distributed as follows: without detectable pre-infection with arenaviruses (n = 54); with pre-existing antibodies to JUN (n = 55); with pre-existing antibodies to LCM (n = 51). The remaining 170 individuals received placebo. Levels of anti-JUN NT Ab displayed a trend in which titers increased with the virulence of the infecting strain, from C#1 (X = 49), through subclinical JUN infection (X = 229), vaccination following subclinical infection (X = 367) to JUN clinical infection (X =773). It was also found that the mean titer of NT Ab to C#1 did not vary significantly during 2 years of study and was: a) significantly lower than that elicited by wild strains of JUN, both clinical and subclinical infections (p < 0.01); b) significantly increased the titers of pre-existing anti-JUN Ab (p < 0.01); and c) was not modified by pre-existing anti-LCM Ab (p > 0.05). These data indicate that the immune response to C#1 boosts pre-existing immunity to JUN virus and is not changed by previous experience with LCM virus.
Subject(s)
Antibodies, Viral/blood , Arenaviridae Infections/immunology , Arenaviruses, New World/immunology , Hemorrhagic Fever, American/prevention & control , Lymphocytic Choriomeningitis/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Adolescent , Adult , Aged , Arenaviridae Infections/virology , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/virology , Humans , Junin virus/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Male , Middle Aged , Neutralization Tests , Treatment Outcome , VaccinationABSTRACT
Tacaribe virus (TACV) is an arenavirus that is genetically and antigenically closely related to Junin virus (JUNV), the aetiological agent of Argentine haemorrhagic fever (AHF). It is well established that TACV protects experimental animals fully against an otherwise lethal challenge with JUNV. To gain information on the nature of the antigens involved in cross-protection, recombinant vaccinia viruses were constructed that express the glycoprotein precursor (VV-GTac) or the nucleocapsid protein (VV-N) of TACV. TACV proteins expressed by vaccinia virus were indistinguishable from authentic virus proteins by gel electrophoresis. Guinea pigs inoculated with VV-GTac or VV-N elicited antibodies that immunoprecipitated authentic TACV proteins. Antibodies generated by VV-GTac neutralized TACV infectivity. Levels of antibodies after priming and boosting with recombinant vaccinia virus were comparable to those elicited in TACV infection. To evaluate the ability of recombinant vaccinia virus to protect against experimental AHF, guinea pigs were challenged with lethal doses of JUNV. Fifty per cent of the animals immunized with VV-GTac survived, whereas all animals inoculated with VV-N or vaccinia virus died. Having established that the heterologous glycoprotein protects against JUNV challenge, a recombinant vaccinia virus was constructed that expresses JUNV glycoprotein precursor (VV-GJun). The size and reactivity to monoclonal antibodies of the vaccinia virus-expressed and authentic JUNV glycoproteins were indistinguishable. Seventy-two per cent of the animals inoculated with two doses of VV-GJun survived lethal JUNV challenge. Protection with either VV-GJun or VV-GTac occurred in the presence of low or undetectable levels of neutralizing antibodies to JUNV.
Subject(s)
Arenaviruses, New World/immunology , Glycoproteins/immunology , Hemorrhagic Fever, American/prevention & control , Junin virus , Viral Proteins/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral/genetics , Antigens, Viral/immunology , Arenaviruses, New World/genetics , Arenaviruses, New World/metabolism , Cell Line , Cross Reactions , Glycoproteins/genetics , Glycoproteins/metabolism , Guinea Pigs , Hemorrhagic Fever, American/virology , Immunization , Junin virus/genetics , Junin virus/immunology , Junin virus/metabolism , Male , Neutralization Tests , Nucleocapsid/genetics , Nucleocapsid/immunology , Nucleocapsid/metabolism , Precipitin Tests , Protein Precursors/genetics , Protein Precursors/immunology , Protein Precursors/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Vaccinia virus/genetics , Vaccinia virus/immunology , Vaccinia virus/metabolismABSTRACT
Despite intensive surveillance, Venezuelan hemorrhagic fever (VHF), caused by Guanarito (GTO) virus, has been detected in only a small region of western Venezuela. To determine whether VHF is associated with a particular regional GTO virus strain(s), 29 isolates from rodents and humans throughout the surrounding regions were analyzed by partial sequencing of the nucleocapsid protein gene. Phylogenetic trees delineated nine distinct GTO genotypes that differ by 4-17% in nucleotides and up to 9% in amino acid sequences; most appeared to be restricted to discrete geographic regions, although a few genotypes were isolated in several locations. Each genotype included at least one strain recovered from a rodent, but only two genotypes were isolated from VHF cases. The presence outside of the endemic/epidemic region of two genotypes isolated also from VHF cases suggests that human pathogenic viruses occur outside of the endemic zone, but do not frequently infect people and/or cause apparent disease there. VHF does not appear to be associated with a GTO virus genotype that is restricted to a certain rodent species. When quasispecies diversity was examined, rodent isolates had higher sequence variation than human isolates. One rodent isolate included a mixture of two phylogenetically distinct genotypes, suggesting a dual infection.
Subject(s)
Arenaviruses, New World/classification , Arenaviruses, New World/genetics , Genes, Viral , Hemorrhagic Fever, American/virology , Rodentia/virology , Animals , Arenaviruses, New World/immunology , Arenaviruses, New World/isolation & purification , Endemic Diseases , Genetic Variation , Genotype , Hemorrhagic Fever, American/epidemiology , Hemorrhagic Fever, American/veterinary , Humans , Molecular Sequence Data , Nucleocapsid/genetics , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Rodent Diseases/virology , Sequence Analysis, DNA , Venezuela/epidemiologyABSTRACT
Argentine hemorrhagic fever (AHF), caused by the arenavirus Junin, is a major public health problem among agricultural workers in Argentina. A prospective, randomized, double-blind, placebo-controlled, efficacy trial of Candid 1, a live attenuated Junin virus vaccine, was conducted over two consecutive epidemic seasons among 6500 male agricultural workers in the AHF-endemic region. Twenty-three men developed laboratory-confirmed AHF during the study; 22 received placebo and 1 received vaccine (vaccine efficacy 95%; 95% confidence interval [CI], 82%-99%). Three additional subjects in each group developed laboratory-confirmed Junin virus infection associated with mild illnesses that did not fulfill the clinical case definition for AHF, yielding a protective efficacy for prevention of any illness associated with Junin virus infection of 84% (95% CI, 60%-94%). No serious adverse events were attributed to vaccination. Candid 1, the first vaccine for the prevention of illness caused by an arenavirus, is safe and highly efficacious.
Subject(s)
Arenaviruses, New World/immunology , Hemorrhagic Fever, American/prevention & control , Hemorrhagic Fever, American/therapy , Vaccines, Attenuated/therapeutic use , Viral Vaccines/therapeutic use , Adolescent , Adult , Agricultural Workers' Diseases/prevention & control , Agricultural Workers' Diseases/therapy , Agricultural Workers' Diseases/virology , Animals , Antibodies, Viral/analysis , Antibodies, Viral/immunology , Arenaviruses, New World/growth & development , Argentina , Cells, Cultured , Chlorocebus aethiops , Double-Blind Method , Hemorrhagic Fever, American/diagnosis , Humans , Male , Middle Aged , Prospective Studies , Seasons , Vaccines, Attenuated/adverse effects , Vero Cells , Viral Vaccines/adverse effectsABSTRACT
Bolivian hemorrhagic fever (BHF) is a potentially severe febrile illness caused by Machupo virus (family Arenaviridae). Initial symptoms include headache, fever, arthralgia, and myalgia. In the later stages of this illness, patients may develop hemorrhagic manifestations including subconjunctival hemorrhage, epistaxis, hematemesis, melena, and hematuria, as well as neurological signs including tremor, seizures, and coma. During the BHF epidemics of the 1960s, convalescent-phase immune plasma from survivors of BHF was administered to selected patients infected with Machupo virus. However, there is currently a paucity of survivors of BHF who can donate immune plasma, and there is no active program for collection and storage of BHF immune plasma; therefore, we had the opportunity to offer intravenous ribavirin to two of three patients with this potentially life-threatening infection. One patient with laboratory-confirmed Machupo virus infection who received ribavirin recovered without sequelae, as did a second patient with suspected BHF whose epidemiological and clinical features were similar to those of the first patient. This report describes the first use of intravenous ribavirin therapy for BHF in humans, and the results suggest the need for more extensive clinical studies to assess the usefulness of ribavirin for treating BHF.
Subject(s)
Antiviral Agents/therapeutic use , Hemorrhagic Fever, American/drug therapy , Ribavirin/therapeutic use , Adult , Antigens, Viral/analysis , Arenaviruses, New World/immunology , Arenaviruses, New World/isolation & purification , Fatal Outcome , Hemorrhagic Fever, American/physiopathology , Hemorrhagic Fever, American/virology , Humans , Injections, Intravenous , Male , Middle AgedABSTRACT
Immunization of BALB/c, C57BL/6, CBA/calac mice with strain XJ44 of Argentine hemorrhagic fever resulted in changes of nonspecific immunity parameters, such as interferon, interleukin-1, tumor necrosis factor, and natural killers. The formation of a specific humoral and cellular immune response in BALB/c mice immunized with this strain has been demonstrated. Immunization of BALB/c mice with strain XJ44 protected the animals from infection with a heterogeneous strain Carvallo of Bolivian hemorrhagic fever.
Subject(s)
Arenaviruses, New World/immunology , Hemorrhagic Fever, American/prevention & control , Vaccines, Attenuated/therapeutic use , Viral Vaccines/therapeutic use , Animals , Antibodies, Viral/analysis , Cross Reactions , Hemorrhagic Fever, American/immunology , Immunity, Innate , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Neutralization TestsABSTRACT
This paper characterizes Guanarito virus, the etiologic agent of Venezuelan hemorrhagic fever. Based on its morphology and antigenic properties, Guanarito virus appears to be a new member of the Tacaribe complex of the genus Arenavirus, family Arenaviridae. Complement fixation and indirect fluorescent antibody tests showed that Guanarito virus and its antiserum are broadly cross-reactive with other members of the Tacaribe complex, but it can be differentiated from other members of the complex by neutralization test. Guanarito virus causes mortality in suckling mice and adult guinea pigs, but not in adult mice. Inoculated rhesus monkeys developed viremia and became ill; however, they subsequently recovered and responded with production of antibody. To date, all isolates of Guanarito virus have come from sick persons or wild rodents living within a single geographic focus in the central plains of Venezuela.
Subject(s)
Arenaviruses, New World/pathogenicity , Hemorrhagic Fever, American/microbiology , Adult , Animals , Animals, Suckling , Antigens, Viral/analysis , Antigens, Viral/immunology , Arenaviruses, New World/immunology , Arenaviruses, New World/ultrastructure , Cell Line , Complement Fixation Tests , Cross Reactions , Cytopathogenic Effect, Viral , Fluorescent Antibody Technique , Guinea Pigs , Humans , Macaca mulatta , Male , Mice , Microscopy, Electron , Neutralization Tests , Venezuela , Vero Cells , Viremia/microbiologyABSTRACT
During February 1992, field studies on the epidemiology of Venezuelan hemorrhagic fever (VHF) were carried out in a rural area of Portuguesa State in central Venezuela. The objective of this work was to determine the prevalence of infection with Guanarito virus, the etiologic agent of VHF, among wild rodents and humans living within an endemic focus of the disease. A total of 234 rodents, representing nine different species, were collected and their spleens were cultured for virus. Thirty-one Guanarito virus isolates were made from two rodent species: 19 from 40 Sigmodon alstoni and 12 from 106 Zygodontomys brevicauda. Guanarito virus antibody rates among these two species were 5.1% and 15.0%, respectively. Nine of the 12 Z. brevicauda that yielded virus from their spleens also had Guanarito virus antibodies in their sera. In contrast, none of the 19 Guanarito virus-positive S. alstoni had antibodies to the virus. These data suggest that S. alstoni usually develops a persistent nonimmunizing infection with Guanarito virus, while Z. brevicauda develops an immunizing infection. Based on knowledge of the behavior of other human pathogenic arenaviruses, these results imply that S. alstoni is the principal rodent reservoir of Guanarito virus in nature. To determine the prevalence of Guanarito virus infection among humans in the same region, 195 people living near one of the rodent collecting sites were bled and their sera were tested for antibodies to the virus. Five individuals (2.6%) had Guanarito virus antibodies; all were adults, and two had been diagnosed previously as having VHF.(ABSTRACT TRUNCATED AT 250 WORDS)
Subject(s)
Arenaviruses, New World/isolation & purification , Disease Reservoirs , Hemorrhagic Fever, American/epidemiology , Sigmodontinae/microbiology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Viral/blood , Arenaviruses, New World/immunology , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Hemorrhagic Fever, American/transmission , Humans , Male , Middle Aged , Prevalence , Rural Health , Spleen/microbiology , Venezuela/epidemiologyABSTRACT
The safety and immunogenicity of Candid #1, a live-attenuated Junin virus vaccine, were evaluated in rhesus macaques. Candid #1 was inoculated subcutaneously in graded doses ranging from 16 to 127,200 plaque-forming units (PFU) into four groups of five animals each; four controls received saline. There was no significant effect of the immunization on any physical, hematologic, or biochemical parameter measured. Junin virus was recovered by cocultivation from peripheral blood mononuclear cells (PBMC) of 14 (70%) of 20 animals from 1 to 21 days after immunization; 27 (12%) of 223 PBMC samples that represented animals in all four dose groups were positive. In contrast, virus was recovered from the plasma of only two of 20 macaques (two of 225 samples [0.9%]), and only once (by amplification) from throat swabs. No evidence of reversion was detected in any blood isolate. All animals developed a detectable neutralizing antibody response following vaccination. These results indicate that Candid #1 is safe and immunogenic in nonhuman primates.