Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Mar Drugs ; 19(10)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34677476

ABSTRACT

The Hantaan orthohantavirus (genovariant Amur-AMRV) is a rodent-borne zoonotic virus; it is the causative agent of haemorrhagic fever with renal syndrome in humans. The currently limited therapeutic options require the development of effective anti-orthohantavirus drugs. The ability of native fucoidan from Fucus evanescens (FeF) and its enzymatically prepared high-molecular-weight (FeHMP) and low-molecular-weight (FeLMP) fractions to inhibit different stages of AMRV infection in Vero cells was studied. The structures of derivatives obtained were determined using nuclear magnetic resonance (NMR) spectroscopy. We found that fucoidan and its derivatives exhibited significant antiviral activity by affecting the early stages of the AMRV lifecycle, notably virus attachment and penetration. The FeHMP and FeLMP fractions showed the highest anti-adsorption activity by inhibiting AMRV focus formation, with a selective index (SI) > 110; FeF had an SI of ~70. The FeLMP fraction showed a greater virucidal effect compared with FeF and the FeHMP fraction. It was shown by molecular docking that 2O-sulphated fucotetrasaccharide, a main component of the FeLMP fraction, is able to bind with the AMRV envelope glycoproteins Gn/Gc and with integrin ß3 to prevent virus-cell interactions. The relatively small size of these sites of interactions explains the higher anti-AMRV activity of the FeLMP fraction.


Subject(s)
Antiviral Agents/pharmacology , Orthohantavirus/drug effects , Phaeophyceae , Polysaccharides/pharmacology , Animals , Antiviral Agents/chemistry , Aquatic Organisms , Humans , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Weight , Polysaccharides/chemistry
2.
Viruses ; 13(4)2021 04 16.
Article in English | MEDLINE | ID: mdl-33923413

ABSTRACT

Hemorrhagic fever viruses, among them orthohantaviruses, arenaviruses and filoviruses, are responsible for some of the most severe human diseases and represent a serious challenge for public health. The current limited therapeutic options and available vaccines make the development of novel efficacious antiviral agents an urgent need. Inhibiting viral attachment and entry is a promising strategy for the development of new treatments and to prevent all subsequent steps in virus infection. Here, we developed a fluorescence-based screening assay for the identification of new antivirals against hemorrhagic fever virus entry. We screened a phytochemical library containing 320 natural compounds using a validated VSV pseudotype platform bearing the glycoprotein of the virus of interest and encoding enhanced green fluorescent protein (EGFP). EGFP expression allows the quantitative detection of infection and the identification of compounds affecting viral entry. We identified several hits against four pseudoviruses for the orthohantaviruses Hantaan (HTNV) and Andes (ANDV), the filovirus Ebola (EBOV) and the arenavirus Lassa (LASV). Two selected inhibitors, emetine dihydrochloride and tetrandrine, were validated with infectious pathogenic HTNV in a BSL-3 laboratory. This study provides potential therapeutics against emerging virus infection, and highlights the importance of drug repurposing.


Subject(s)
Antiviral Agents/therapeutic use , Drug Evaluation, Preclinical/methods , Hantavirus Infections/drug therapy , Orthohantavirus/drug effects , Virus Internalization/drug effects , Humans
3.
s.l; Ministerio de Salud de Rio Negro; 2021. 6 p.
Non-conventional in Spanish | BIGG - GRADE guidelines | ID: biblio-1281955

ABSTRACT

El panel priorizó la posibilidad de una reducción significativa en la mortalidad por sobre la incertidumbre en los efectos de la intervención (muy baja certeza en la evidencia), los efectos adversos del tratamiento y la carga del tratamiento en términos de utilización de recursos necesarios para implementar la intervención. La evidencia en estudios en humanos con infección por hantavirus con síndrome cardiopulmonar muestra que el tratamiento con ribavirina en humanos podria reducir el riesgo de muerte en etapa asintomatica o prodromica (RR 0.28; IC 95%: 0,08 a 1). En ensayos en modelos animales infectados con hantavirus en fase asintomatica (hasta 7 dias luego de la inoculacion del virus) mostró una disminución de la mortalidad entre el 12% y el 81% (RR 0.41; 0.19 a 0.88. Sin embargo, no es posible determinar el impacto sobre la mortalidad, ya que la certeza en la evidencia sobre este desenlace resultó muy baja por lo que existe incertidumbre sobre el efecto de ribavirina en pacientes con infección por hantavirus en etapas asintomática o prodrómica. La incidencia de efectos adversos que llevaron a discontinuar el tratamiento fue de 4%. El efecto adverso más frecuentemente reportado fue anemia hemolítica con una incidencia de 14% (la mayoría de los casos no fueron severos). Otros efectos adversos menos frecuentes fueron nefrotoxicidad, pancreatitis, rash y hepatotoxicidad. Existe una incertidumbre en relación al riesgo de efectos adversos severos asociados a la ribavirina dada la muy baja certeza de la evidencia, que se basa en estudios observacionales. La ribavirina probablemente se asocie con anemia con un 57.8% más (25.3 más a 111.9 más) en la rama intervención.


Subject(s)
Humans , Orthohantavirus/drug effects , Hantavirus Infections/drug therapy , Ribavirin/therapeutic use , Enzyme-Linked Immunosorbent Assay/methods , Carrier State , Polymerase Chain Reaction/methods , Orthohantavirus/isolation & purification , Hantavirus Infections/diagnosis
4.
Front Cell Infect Microbiol ; 10: 561502, 2020.
Article in English | MEDLINE | ID: mdl-33251157

ABSTRACT

Andes virus (ANDV) and Sin Nombre virus (SNV), highly pathogenic hantaviruses, cause hantavirus pulmonary syndrome in the Americas. Currently no therapeutics are approved for use against these infections. Griffithsin (GRFT) is a high-mannose oligosaccharide-binding lectin currently being evaluated in phase I clinical trials as a topical microbicide for the prevention of human immunodeficiency virus (HIV-1) infection (ClinicalTrials.gov Identifiers: NCT04032717, NCT02875119) and has shown broad-spectrum in vivo activity against other viruses, including severe acute respiratory syndrome coronavirus, hepatitis C virus, Japanese encephalitis virus, and Nipah virus. In this study, we evaluated the in vitro antiviral activity of GRFT and its synthetic trimeric tandemer 3mGRFT against ANDV and SNV. Our results demonstrate that GRFT is a potent inhibitor of ANDV infection. GRFT inhibited entry of pseudo-particles typed with ANDV envelope glycoprotein into host cells, suggesting that it inhibits viral envelope protein function during entry. 3mGRFT is more potent than GRFT against ANDV and SNV infection. Our results warrant the testing of GRFT and 3mGRFT against ANDV infection in animal models.


Subject(s)
Antiviral Agents/pharmacology , Hantavirus Pulmonary Syndrome/virology , Lectins/pharmacology , Orthohantavirus/drug effects , Sin Nombre virus/drug effects , Cell Culture Techniques , Cell Line , Orthohantavirus/physiology , Hantavirus Pulmonary Syndrome/drug therapy , Humans , Sin Nombre virus/physiology
5.
Microbes Infect ; 22(6-7): 272-277, 2020.
Article in English | MEDLINE | ID: mdl-32445882

ABSTRACT

Hantavirus (HV), a pathogen of animal infectious diseases that poses a threat to humans, has attracted extensive attention. Clinically, HV can cause hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS), between which HFRS is mostly in Eurasia, and HPS is mostly in the Americas. This paper reviews the research progress of small-molecule inhibitors of HV.


Subject(s)
Antiviral Agents/pharmacology , Orthohantavirus/drug effects , Small Molecule Libraries/pharmacology , Animals , Orthohantavirus/physiology , Hantavirus Infections/virology , Host-Pathogen Interactions/drug effects , Humans , Peptides/chemistry , Peptides/pharmacology , Viral Proteins/antagonists & inhibitors , Viral Proteins/chemistry , Virus Replication/drug effects
6.
Viruses ; 12(1)2020 01 20.
Article in English | MEDLINE | ID: mdl-31968537

ABSTRACT

Orthohantaviruses, previously known as hantaviruses (family Hantaviridae, order Bunyavirales), are emerging zoonoses hosted by different rodent and insectivore species. Orthohantaviruses are transmitted by aerosolized excreta (urine, saliva and feces) of their reservoir hosts. When transmitted to humans, they cause hemorrhagic fever with renal syndrome (HFRS) in Asia and Europe and hantavirus (cardio) pulmonary syndrome (HPS) in the Americas. Clinical studies have shown that early treatments of HFRS patients with ribavirin (RBV) improve prognosis. Nevertheless, there is the need for urgent development of specific antiviral drugs. In the search for new RNA virus inhibitors, we recently identified a series of variously substituted 5,6-dichloro-1(2)-phenyl-1(2)H-benzo[d][1,2,3]triazole derivatives active against the human respiratory syncytial virus (HRSV). Interestingly, several 2-phenyl-benzotriazoles resulted in fairly potent inhibitors of the Hantaan virus in a chemiluminescence focus reduction assay (C-FRA) showing an EC50 = 4-5 µM, ten-fold more active than ribavirin. Currently, there are no FDA approved drugs for the treatment of orthohantavirus infections. Antiviral activities and cytotoxicity profiles suggest that 5,6-dichloro-1(2)-phenyl-1(2)H-benzo[d][1,2,3]triazoles could be promising candidates for further investigation as a potential treatment of hantaviral diseases.


Subject(s)
Antiviral Agents/pharmacology , Orthohantavirus/drug effects , Triazoles/chemistry , Triazoles/pharmacology , Animals , Antiviral Agents/chemistry , Chlorocebus aethiops , Drug Discovery , Vero Cells
7.
Enferm Infecc Microbiol Clin (Engl Ed) ; 37(9): 602-608, 2019 Nov.
Article in English, Spanish | MEDLINE | ID: mdl-29907366

ABSTRACT

Ribavirin is a molecule with antiviral activity against different viruses. In clinical practice, it has made its niche almost exclusively for the treatment of the hepatitisC virus. However, there are other diseases in which it could be of benefit and it has the advantage of being suitable for oral, intravenous and inhaled administration. We conducted a review of the indications of the main drug agencies (Spanish, European and American) and other possible indications, mainly haemorrhagic fevers and coronavirus.


Subject(s)
Antiviral Agents/therapeutic use , Ribavirin/therapeutic use , Virus Diseases/drug therapy , Viruses/drug effects , Adenoviridae Infections/drug therapy , Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Arenaviruses, New World/drug effects , Clinical Trials as Topic , Coronavirus Infections/drug therapy , Orthohantavirus/drug effects , Hantavirus Infections/drug therapy , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever, American/drug therapy , Hemorrhagic Fever, Crimean/drug therapy , Humans , Lassa Fever/drug therapy , Lassa virus/drug effects , Meta-Analysis as Topic , Middle East Respiratory Syndrome Coronavirus/drug effects , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects
8.
Sci Transl Med ; 10(468)2018 11 21.
Article in English | MEDLINE | ID: mdl-30463919

ABSTRACT

Andes hantavirus (ANDV) is an etiologic agent of hantavirus cardiopulmonary syndrome (HCPS), a severe disease characterized by fever, headache, and gastrointestinal symptoms that may progress to hypotension, pulmonary failure, and cardiac shock that results in a 25 to 40% case-fatality rate. Currently, there is no specific treatment or vaccine; however, several studies have shown that the generation of neutralizing antibody (Ab) responses strongly correlates with survival from HCPS in humans. In this study, we screened 27 ANDV convalescent HCPS patient sera for their capacity to bind and neutralize ANDV in vitro. One patient who showed high neutralizing titer was selected to isolate ANDV-glycoprotein (GP) Abs. ANDV-GP-specific memory B cells were single cell sorted, and recombinant immunoglobulin G antibodies were cloned and produced. Two monoclonal Abs (mAbs), JL16 and MIB22, potently recognized ANDV-GPs and neutralized ANDV. We examined the post-exposure efficacy of these two mAbs as a monotherapy or in combination therapy in a Syrian hamster model of ANDV-induced HCPS, and both mAbs protected 100% of animals from a lethal challenge dose. These data suggest that monotherapy with mAb JL16 or MIB22, or a cocktail of both, could be an effective post-exposure treatment for patients infected with ANDV-induced HCPS.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hantavirus Infections/drug therapy , Hantavirus Infections/prevention & control , Orthohantavirus/physiology , Recombinant Proteins/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , B-Lymphocytes/drug effects , Glycoproteins/immunology , HEK293 Cells , Orthohantavirus/drug effects , Hantavirus Infections/blood , Hantavirus Infections/immunology , Humans , Immunologic Memory/drug effects , Recombinant Proteins/pharmacology , Survivors
9.
SLAS Discov ; 23(7): 634-645, 2018 08.
Article in English | MEDLINE | ID: mdl-29608398

ABSTRACT

Hantaviruses cause hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS), which infects more than 200,000 people worldwide. Sin Nombre virus (SNV) and Andes virus (ANDV) cause the most severe form of HCPS, with case fatality ratios of 30%-40%. There are no specific therapies or vaccines for SNV. Using high-throughput flow cytometry, we screened the Prestwick Chemical Library for small-molecule inhibitors of the binding interaction between UV-inactivated and fluorescently labeled SNVR18 particles, and decay-accelerating factor (DAF) expressed on Tanoue B cells. Eight confirmed hit compounds from the primary screen were investigated further in secondary screens that included infection inhibition, cytotoxicity, and probe interference. Antimycin emerged as a bona fide hit compound that inhibited cellular infection of the major HCPS (SNV)- and HCPS (Hantaan)-causing viruses. Confirming our assay's ability to detect active compounds, orthogonal testing of the hit compound showed that antimycin binds directly to the virus particle and blocks recapitulation of physiologic integrin activation caused by SNV binding to the integrin PSI domain.


Subject(s)
Antiviral Agents/pharmacology , Flow Cytometry , High-Throughput Screening Assays , Orthohantavirus/drug effects , Virus Internalization/drug effects , Animals , Biomarkers , Cell Survival/drug effects , Chlorocebus aethiops , Dose-Response Relationship, Drug , Flow Cytometry/methods , Orthohantavirus/physiology , Hantavirus Infections/drug therapy , Hantavirus Infections/virology , Humans , Models, Biological , Reproducibility of Results , Vero Cells
10.
Antiviral Res ; 147: 107-115, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29017779

ABSTRACT

Hantavirus (HV) infection, which underlies hantavirus hemorrhagic fever with renal syndrome and hantavirus pulmonary syndrome, remains to be a severe clinical challenge. Here, we synthesized small interfering RNAs (siRNAs) that target the encoding sequences of HV strain 76-118, and validated their inhibitory role in virus replication in HV-infected monkey kidney Vero E6 cells. A chimeric protein, 3G1-Cκ-tP, consisting of a single-chain antibody fragment (3G1) against the HV surface envelop glycoprotein, the constant region of human immunoglobulin κ chain (Cκ), and truncated protamine (amino acids 8-29, tP), was further generated. The fusion protein showed high affinity to HV antigen on the infected cell membrane, and internalized through clathrin-mediated endocytosis; it bound to siRNAs via the basic nucleic acid-rich protamine fragment, leading to their specific delivery into HV-infected cells and efficient inhibition of virus replication. An encephalitis mouse model was established via intracranial HV administration. Intraperitoneal injection of siRNAs complexed with 3G1-Cκ-tP achieved specific distribution of siRNAs in HV-infected brain cells, significantly reduced HV antigen levels, and effective protection from HV infection-derived animal death. These results provide a compelling rationale for novel therapeutic protocols designed for HV infection and related disorders.


Subject(s)
Drug Delivery Systems , Hemorrhagic Fever with Renal Syndrome/virology , Orthohantavirus/drug effects , RNA, Small Interfering/pharmacology , Virus Replication/drug effects , Animals , Animals, Newborn , Antigens, Viral/metabolism , Chlorocebus aethiops , Disease Models, Animal , Hemorrhagic Fever with Renal Syndrome/drug therapy , Hemorrhagic Fever with Renal Syndrome/pathology , Humans , Mice , Mice, Inbred BALB C , Protein Binding , RNA Interference , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Single-Chain Antibodies/genetics , Vero Cells , Viral Envelope Proteins/genetics , Viral Load/drug effects
11.
SLAS Discov ; 22(6): 767-774, 2017 07.
Article in English | MEDLINE | ID: mdl-28340538

ABSTRACT

Humans acquire hantavirus infection by the inhalation of aerosolized excreta of infected rodent hosts. There is no treatment for hantavirus diseases at present. Therapeutic intervention during early stages of viral infection can improve the outcome of this zoonotic viral illness. The interaction between an evolutionary conserved sequence at the 5' terminus of hantaviral genomic RNA and hantavirus nucleocapsid protein plays a critical role in the hantavirus replication cycle. This unique interaction is a novel target for therapeutic intervention of hantavirus disease. We developed a very sensitive, tractable, and cost-effective fluorescence-based assay to monitor the interaction between the nucleocapsid protein and the target RNA sequence. The assay was optimized for high-throughput screening of chemical libraries to identify molecules that interrupt this RNA-protein interaction. The assay was validated using a library of 6880 chemical compounds. This validation screen demonstrated the reproducibility and validity of required statistical criteria for high-throughput screening. The assay is ready to use for high-throughput screening of large chemical libraries to identify antihantaviral therapeutic molecules and can be amenable to similar targets in other viruses.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , High-Throughput Screening Assays , Orthohantavirus/drug effects , Drug Discovery/methods , Orthohantavirus/isolation & purification , Orthohantavirus/physiology , Hantavirus Infections/drug therapy , Hantavirus Infections/virology , Humans , Protein Binding , RNA, Viral/genetics , RNA, Viral/metabolism , Reproducibility of Results , Untranslated Regions , Viral Proteins/metabolism , Virus Replication/drug effects
12.
Antiviral Res ; 132: 66-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27233645

ABSTRACT

Hantavirus pulmonary syndrome (HPS) is a severe disease caused by hantavirus infection of pulmonary microvascular endothelial cells leading to microvascular leakage, pulmonary edema, pleural effusion and high case fatality. Previously, we demonstrated that Andes virus (ANDV) infection caused up-regulation of vascular endothelial growth factor (VEGF) and concomitant downregulation of the cellular adhesion molecule VE-cadherin leading to increased permeability. Analyses of human HPS-patient sera have further demonstrated increased circulating levels of VEGF. Here we investigate the impact of a small molecule antagonist of the VEGF receptor 2 (VEGFR-2) activation in vitro, and overall impact on survival in the Syrian hamster model of HPS.


Subject(s)
Hantavirus Pulmonary Syndrome/virology , Orthohantavirus/drug effects , Orthohantavirus/physiology , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Animals , Capillary Permeability/drug effects , Cricetinae , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/virology , Hantavirus Pulmonary Syndrome/metabolism , Hantavirus Pulmonary Syndrome/mortality , Phosphorylation/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Viral Load
13.
Food Environ Virol ; 8(2): 120-4, 2016 06.
Article in English | MEDLINE | ID: mdl-26943130

ABSTRACT

The effect of carrageenans and fucoidans on the activity of Hantavirus is studied. It has been found that among carrageenans a significant antiviral effect is exerted by the ι-type, which decreases the viral titer by 2.5 log focus forming units per mL; among fucoidans, by a preparation from Laminaria cichorioides, which reduces the number of infected cells from 27.0 to 5.3 after pretreatment of both the macrophage culture and Hantavirus. The antiviral effect of fucoidan from Laminaria japonica is shown to grow in direct proportion to the increase of dose of the preparation.


Subject(s)
Carrageenan/pharmacology , Laminaria/chemistry , Orthohantavirus/drug effects , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Carrageenan/isolation & purification , Orthohantavirus/physiology , Plant Extracts/isolation & purification , Polysaccharides/isolation & purification , Seaweed/chemistry
14.
Antiviral Res ; 126: 62-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26711718

ABSTRACT

Favipiravir is approved in Japan to treat novel or re-emerging influenza viruses, and is active against a broad spectrum of RNA viruses, including Ebola. Ribavirin is the only other licensed drug with activity against multiple RNA viruses. Recent studies show that ribavirin and favipiravir act synergistically to inhibit bunyavirus infections in cultured cells and laboratory mice, likely due to their different mechanisms of action. Convalescent immune globulin is the only approved treatment for Argentine hemorrhagic fever caused by the rodent-borne Junin arenavirus. We previously reported that favipiravir is highly effective in a number of small animal models of Argentine hemorrhagic fever. We now report that addition of low dose of ribavirin synergistically potentiates the activity of favipiravir against Junin virus infection of guinea pigs and another arenavirus, Pichinde virus infection of hamsters. This suggests that the efficacy of favipiravir against hemorrhagic fever viruses can be further enhanced through the addition of low-dose ribavirin.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Hemorrhagic Fevers, Viral/drug therapy , Pyrazines/pharmacology , RNA Viruses/drug effects , Ribavirin/pharmacology , Animals , Arenavirus/drug effects , Chlorocebus aethiops , Cricetinae , Dengue Virus/drug effects , Disease Models, Animal , Drug Synergism , Female , Guinea Pigs , Orthohantavirus/drug effects , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever, American/drug therapy , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fevers, Viral/blood , Hemorrhagic Fevers, Viral/veterinary , Hemorrhagic Fevers, Viral/virology , Junin virus/drug effects , Male , Mesocricetus , Mice , Vero Cells
15.
Antivir Ther ; 20(4): 377-86, 2015.
Article in English | MEDLINE | ID: mdl-25316807

ABSTRACT

BACKGROUND: In Chile, Andes virus (ANDV) is the sole aetiological agent of hantavirus cardiopulmonary syndrome (HCPS) with mean annual incidence of 55 cases, 32% case fatality rate (CFR) and no specific treatment. Neutralizing antibody (NAb) titres at hospital admission correlate inversely with HCPS severity. We designed an open trial to explore safety and efficacy and evaluate pharmacokinetics of immune plasma as a treatment strategy for this disease. METHODS: We performed plasmapheresis on donors at least 6 months after HCPS and measured NAb titres through a focus-reduction neutralization test. Subjects admitted to 10 study sites with suspected/confirmed HCPS were eligible for treatment with immune plasma by intravenous infusion at an ANDV NAb dose of 5,000 U/kg. HCPS was confirmed through immunoglobulin M serology or reverse transcriptase-PCR. The main outcome was mortality within 30 days. RESULTS: From 2008-2012, we enrolled and treated 32 cases and confirmed HCPS in 29. CFR of hantavirus plasma-treated cases was 4/29 (14%); CFR of non-treated cases in the same period in Chile was 63/199 (32%; P=0.049, OR=0.35, CI=0.12, 0.99); CFR of non-treated cases at the same study sites between 2005-2012 was 18/66 (27%; (P=0.15, OR=0.43, CI=0.14, 1.34) and CFR in a previous methylprednisolone treatment study was 20/60 (33%; P=0.052, OR=0.32, CI=0.10, 1.00). We detected no serious adverse events associated to plasma infusion. Plasma NAb titres reached in recipients were variable and viral load remained stable. CONCLUSIONS: Human ANDV immune plasma infusion appears safe for HCPS. We observed a decrease in CFR in treated cases with borderline significance that will require further studies for confirmation.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Hantavirus Infections/therapy , Immune Sera/pharmacology , RNA, Viral/antagonists & inhibitors , Adult , Female , Glucocorticoids/therapeutic use , Orthohantavirus/drug effects , Orthohantavirus/growth & development , Orthohantavirus/immunology , Hantavirus Infections/immunology , Hantavirus Infections/mortality , Hantavirus Infections/virology , Heart/drug effects , Heart/physiopathology , Heart/virology , Humans , Lung/drug effects , Lung/immunology , Lung/pathology , Lung/virology , Male , Methylprednisolone/therapeutic use , Middle Aged , Neutralization Tests , Plasmapheresis , RNA, Viral/blood , RNA, Viral/immunology , Severity of Illness Index , Survival Analysis , Syndrome , Viral Load/drug effects
16.
PLoS One ; 9(6): e99764, 2014.
Article in English | MEDLINE | ID: mdl-24924189

ABSTRACT

Andes virus (ANDV) is the most common causative agent of hantavirus pulmonary syndrome (HPS) in the Americas, and is the only hantavirus associated with human-to-human transmission. Case fatality rates of ANDV-induced HPS are approximately 40%. There are currently no effective vaccines or antivirals against ANDV. Since HPS severity correlates with viral load, we tested small interfering RNA (siRNA) directed against ANDV genes as a potential antiviral strategy. We designed pools of 4 siRNAs targeting each of the ANDV genome segments (S, M, and L), and tested their efficacy in reducing viral replication in vitro. The siRNA pool targeting the S segment reduced viral transcription and replication in Vero-E6 cells more efficiently than those targeting the M and L segments. In contrast, siRNAs targeting the S, M, or L segment were similar in their ability to reduce viral replication in human lung microvascular endothelial cells. Importantly, these siRNAs inhibit ANDV replication even if given after infection. Taken together, our findings indicate that siRNAs targeting the ANDV genome efficiently inhibit ANDV replication, and show promise as a strategy for developing therapeutics against ANDV infection.


Subject(s)
Orthohantavirus/drug effects , Orthohantavirus/physiology , RNA, Small Interfering/pharmacology , Virus Replication/drug effects , Animals , Cells, Cultured , Chlorocebus aethiops , Endothelial Cells/drug effects , Endothelial Cells/virology , Orthohantavirus/genetics , Hantavirus Pulmonary Syndrome/pathology , Hantavirus Pulmonary Syndrome/virology , Humans , Lung/cytology , Lung/drug effects , Lung/pathology , Lung/virology , Primary Cell Culture , RNA, Viral/antagonists & inhibitors , RNA, Viral/genetics , Vero Cells , Virus Replication/genetics
17.
Viruses ; 5(11): 2704-20, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24217424

ABSTRACT

Andes virus, ANDV, harbored by wild rodents, causes the highly lethal hantavirus pulmonary syndrome (HPS) upon transmission to humans resulting in death in 30% to 50% of the cases. As there is no treatment for this disease, we systematically tested the efficacy of ribavirin in vitro and in an animal model. In vitro assays confirmed antiviral activity and determined that the most effective doses were 40 µg/mL and above. We tested three different concentrations of ribavirin for their capability to prevent HPS in the ANDV hamster model following an intranasal challenge. While the highest level of ribavirin (200 mg/kg) was toxic to the hamster, both the middle (100 mg/kg) and the lowest concentration (50 mg/kg) prevented HPS in hamsters without toxicity. Specifically, 8 of 8 hamsters survived intranasal challenge for both of those groups whereas 7 of 8 PBS control-treated animals developed lethal HPS. Further, we report that administration of ribavirin at 50 mg/kg/day starting on days 6, 8, 10, or 12 post-infection resulted in significant protection against HPS in all groups. Administration of ribavirin at 14 days post-infection also provided a significant level of protection against lethal HPS. These data provide in vivo evidence supporting the potential use of ribavirin as a post-exposure treatment to prevent HPS after exposure by the respiratory route.


Subject(s)
Antiviral Agents/administration & dosage , Hantavirus Pulmonary Syndrome/prevention & control , Orthohantavirus/drug effects , Ribavirin/administration & dosage , Animals , Antiviral Agents/adverse effects , Cricetinae , Disease Models, Animal , Female , Orthohantavirus/physiology , Hantavirus Pulmonary Syndrome/drug therapy , Hantavirus Pulmonary Syndrome/virology , Humans , Mesocricetus , Nasal Cavity/virology , Ribavirin/adverse effects
18.
Antimicrob Agents Chemother ; 57(10): 4673-80, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23856782

ABSTRACT

Hantavirus pulmonary syndrome (HPS) is caused by infection with several Sigmodontinae- and Neotominae-borne hantaviruses and has a case fatality rate of 30 to 50%. Humans often become infected by inhalation of materials contaminated with virus-laden rodent urine or saliva, although human-to-human transmission has also been documented for Andes virus (ANDV). The ability to transmit via aerosolization, coupled with the high mortality rates and lack of therapeutic options, makes the development of medical countermeasures against HPS imperative. In the present study, we evaluated the efficacy of the broad-spectrum antiviral agent favipiravir (T-705) against Sin Nombre virus (SNV) and ANDV, the predominant causes of HPS in North and South America, respectively. In vitro, T-705 potently inhibited SNV and ANDV, as evidenced by decreased detection of viral RNA and reduced infectious titers. For both viruses, the 90% effective concentration was estimated at ≤5 µg/ml (≤31.8 µM). In the lethal ANDV hamster model, daily administration of oral T-705 at 50 or 100 mg/kg of body weight diminished the detection of viral RNA and antigen in tissue specimens and significantly improved survival rates. Oral T-705 therapy remained protective against HPS when treatment was initiated prior to the onset of viremia. No disease model for SNV exists; however, using a hamster-adapted SNV, we found that daily administration of oral T-705 significantly reduced the detection of SNV RNA and antigen in tissue specimens, suggesting that the compound would also be effective against HPS in North America. Combined, these results suggest that T-705 treatment is beneficial for postexposure prophylaxis against HPS-causing viruses and should be considered for probable exposures.


Subject(s)
Amides/therapeutic use , Antiviral Agents/therapeutic use , Hantavirus Pulmonary Syndrome/drug therapy , Pyrazines/therapeutic use , Animals , Cricetinae , Female , Orthohantavirus/drug effects , Orthohantavirus/physiology , Sin Nombre virus/drug effects , Sin Nombre virus/physiology , Virus Replication/drug effects
19.
Antivir Ther ; 18(4): 575-84, 2013.
Article in English | MEDLINE | ID: mdl-23300158

ABSTRACT

BACKGROUND: Microbe-induced over-activation of cytokines, especially tumour necrosis factor (TNF)-α, is key to the pathogenesis of hantavirus infection leading to severe inflammation with high mortality rate. Although ribavirin showed promise in inhibiting viral replication in vitro, its clinical efficacy remains controversial. METHODS: Various concentrations of ribavirin were used to determine its effect on cytokine activation in our infectious model system. RESULTS: Ribavirin decreased the virus load and dose-dependently inhibited the accumulation of RANTES messenger RNA in Andes-virus (ANDV)-infected human endothelial cells, but failed to suppress TNF-α-induced activation of RANTES and interleukin-6 in ANDV-inoculated cultures. This report also shows, for the first time, that the deleterious over-stimulation by TNF-α is mediated by nuclear factor-κB, and describes the effect of ribavirin on cytokine production following ANDV infection. CONCLUSIONS: Although highly effective in preventing ANDV replication and suppressing activation of select inflammatory mediators, the therapeutic efficacy of ribavirin is limited due to its inability to fully inhibit cytokine outburst triggered by hantavirus infection.


Subject(s)
Antiviral Agents/pharmacology , Chemokine CCL5/genetics , NF-kappa B/genetics , Orthohantavirus/drug effects , Ribavirin/pharmacology , Tumor Necrosis Factor-alpha/genetics , Animals , Chemokine CCL5/metabolism , Chlorocebus aethiops , Dose-Response Relationship, Drug , Gene Expression Regulation , Orthohantavirus/physiology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/virology , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vero Cells , Viral Load/drug effects
20.
J Virol ; 87(2): 912-22, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23135723

ABSTRACT

Hantavirus pulmonary syndrome (HPS) is a severe respiratory disease characterized by pulmonary edema, with fatality rates of 35 to 45%. Disease occurs following infection with pathogenic New World hantaviruses, such as Andes virus (ANDV), which targets lung microvascular endothelial cells. During replication, the virus scavenges 5'-m(7)G caps from cellular mRNA to ensure efficient translation of viral proteins by the host cell cap-dependent translation machinery. In cells, the mammalian target of rapamycin (mTOR) regulates the activity of host cap-dependent translation by integrating amino acid, energy, and oxygen availability signals. Since there is no approved pharmacological treatment for HPS, we investigated whether inhibitors of the mTOR pathway could reduce hantavirus infection. Here, we demonstrate that treatment with the FDA-approved rapamycin analogue temsirolimus (CCI-779) blocks ANDV protein expression and virion release but not entry into primary human microvascular endothelial cells. This effect was specific to viral proteins, as temsirolimus treatment did not block host protein synthesis. We confirmed that temsirolimus targeted host mTOR complex 1 (mTORC1) and not a viral protein, as knockdown of mTORC1 and mTORC1 activators but not mTOR complex 2 components reduced ANDV replication. Additionally, primary fibroblasts from a patient with tuberous sclerosis exhibited increased mTORC1 activity and increased ANDV protein expression, which were blocked following temsirolimus treatment. Finally, we show that ANDV glycoprotein Gn colocalized with mTOR and lysosomes in infected cells. Together, these data demonstrate that mTORC1 signaling regulates ANDV replication and suggest that the hantavirus Gn protein may modulate mTOR and lysosomal signaling during infection, thus bypassing the cellular regulation of translation.


Subject(s)
Multiprotein Complexes/metabolism , Orthohantavirus/pathogenicity , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Virus Replication , Antiviral Agents/pharmacology , Cells, Cultured , Gene Knockdown Techniques , Orthohantavirus/drug effects , Orthohantavirus/physiology , Humans , Mechanistic Target of Rapamycin Complex 1 , Protein Kinase Inhibitors/pharmacology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL