Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Med Virol ; 95(2): e28548, 2023 02.
Article in English | MEDLINE | ID: mdl-36734067

ABSTRACT

Crimean-Congo haemorrhagic fever (CCHF) is the most widespread tick-borne viral haemorrhagic fever affecting humans, and yet a licensed drug against the virus (CCHFV) is still not available. While several studies have suggested the efficacy of ribavirin against CCHFV, current literature remains inconclusive. In this study, we have utilised next-generation sequencing to investigate the mutagenic effect of ribavirin on the CCHFV genome during clinical disease. Samples collected from CCHF patients receiving ribavirin treatment or supportive care only at Sivas Cumhuriyet University Hospital, Turkey, were analysed. By comparing the frequency of mutations in each group, we found little evidence of an overall mutagenic effect. This suggests that ribavirin, administered at the acute stages of CCHFV infection (at the World Health Organization-recommended dose) is unable to induce lethal mutagenesis that would cause an extinction event in the CCHFV population and reduce viremia.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo , Hemorrhagic Fever, Crimean , Ribavirin , Humans , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever, Crimean/drug therapy , Hemorrhagic Fever, Crimean/epidemiology , High-Throughput Nucleotide Sequencing , Mutation , Ribavirin/therapeutic use
2.
Vox Sang ; 115(3): 146-151, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31930543

ABSTRACT

BACKGROUND: Emerging viruses like severe acute respiratory syndrome coronavirus (SARS-CoV), Crimean-Congo haemorrhagic fever virus (CCHFV) and Nipah virus (NiV) have been identified to pose a potential threat to transfusion safety. In this study, the ability of the THERAFLEX UV-Platelets and THERAFLEX MB-Plasma pathogen inactivation systems to inactivate these viruses in platelet concentrates and plasma, respectively, was investigated. MATERIALS AND METHODS: Blood products were spiked with SARS-CoV, CCHFV or NiV, and then treated with increasing doses of UVC light (THERAFLEX UV-Platelets) or with methylene blue (MB) plus increasing doses of visible light (MB/light; THERAFLEX MB-Plasma). Samples were taken before and after treatment with each illumination dose and tested for residual infectivity. RESULTS: Treatment with half to three-fourths of the full UVC dose (0·2 J/cm2 ) reduced the infectivity of SARS-CoV (≥3·4 log), CCHFV (≥2·2 log) and NiV (≥4·3 log) to the limit of detection (LOD) in platelet concentrates, and treatment with MB and a fourth of the full light dose (120 J/cm2 ) decreased that of SARS-CoV (≥3·1 log), CCHFV (≥3·2 log) and NiV (≥2·7 log) to the LOD in plasma. CONCLUSION: Our study demonstrates that both THERAFLEX UV-Platelets (UVC) and THERAFLEX MB-Plasma (MB/light) effectively reduce the infectivity of SARS-CoV, CCHFV and NiV in platelet concentrates and plasma, respectively.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/radiation effects , Light , Methylene Blue/pharmacology , Nipah Virus/radiation effects , Severe acute respiratory syndrome-related coronavirus/radiation effects , Ultraviolet Rays , Virus Inactivation , Blood Platelets/virology , Blood Transfusion , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Humans , Nipah Virus/drug effects , Plasma/virology , Severe acute respiratory syndrome-related coronavirus/drug effects
3.
J Antimicrob Chemother ; 74(12): 3432-3439, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31369086

ABSTRACT

OBJECTIVES: Recently, ribavirin has been suggested as a therapeutic approach in Crimean-Congo haemorrhagic fever (CCHF) patients; however, there are controversial findings about its efficacy. In the current study, a meta-analysis was systematically performed to assess the effectiveness of ribavirin administration regarding CCHF patient survival and to explore the most important influential parameters for its efficacy. METHODS: All of the outcomes of the clinically studied CCHF patients who were treated with ribavirin were included in the meta-analysis. RESULTS: Overall, 24 studies met our criteria. Although the studies did not have high quality there was no heterogeneity and publication bias across studies. The results indicated that the administration of ribavirin to CCHF patients significantly decreased the mortality rate (by 1.7-fold) compared with those who did not receive this medication. Furthermore, it was found that the prescription of ribavirin in the initial phase of disease was more effective, and a delay in the start of treatment resulted in a 1.6-fold increase in mortality rate. In addition, interventional therapy resulted in an ∼2.3-fold reduction in the mortality rate of those who received ribavirin along with corticosteroids compared with those who were treated with ribavirin monotherapy. CONCLUSIONS: This meta-analysis reveals that ribavirin should be considered as a crucial antiviral drug in the therapeutic approach used for CCHF patients, especially in early phases of the disease. Additionally, it seems that the administration of corticosteroids alongside ribavirin can play an effective role in alleviation of the disease status, particularly in haemorrhagic phases.


Subject(s)
Antiviral Agents/therapeutic use , Hemorrhagic Fever, Crimean/drug therapy , Ribavirin/therapeutic use , Administration, Oral , Adrenal Cortex Hormones/therapeutic use , Controlled Clinical Trials as Topic , Drug Therapy, Combination , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever, Crimean/mortality , Humans , Observational Studies as Topic , Time Factors
4.
PLoS Pathog ; 13(5): e1006372, 2017 May.
Article in English | MEDLINE | ID: mdl-28542609

ABSTRACT

The recent Middle East respiratory syndrome coronavirus (MERS-CoV), Ebola and Zika virus outbreaks exemplify the continued threat of (re-)emerging viruses to human health, and our inability to rapidly develop effective therapeutic countermeasures. Many viruses, including MERS-CoV and the Crimean-Congo hemorrhagic fever virus (CCHFV) encode deubiquitinating (DUB) enzymes that are critical for viral replication and pathogenicity. They bind and remove ubiquitin (Ub) and interferon stimulated gene 15 (ISG15) from cellular proteins to suppress host antiviral innate immune responses. A variety of viral DUBs (vDUBs), including the MERS-CoV papain-like protease, are responsible for cleaving the viral replicase polyproteins during replication, and are thereby critical components of the viral replication cycle. Together, this makes vDUBs highly attractive antiviral drug targets. However, structural similarity between the catalytic cores of vDUBs and human DUBs complicates the development of selective small molecule vDUB inhibitors. We have thus developed an alternative strategy to target the vDUB activity through a rational protein design approach. Here, we report the use of phage-displayed ubiquitin variant (UbV) libraries to rapidly identify potent and highly selective protein-based inhibitors targeting the DUB domains of MERS-CoV and CCHFV. UbVs bound the vDUBs with high affinity and specificity to inhibit deubiquitination, deISGylation and in the case of MERS-CoV also viral replicative polyprotein processing. Co-crystallization studies further revealed critical molecular interactions between UbVs and MERS-CoV or CCHFV vDUBs, accounting for the observed binding specificity and high affinity. Finally, expression of UbVs during MERS-CoV infection reduced infectious progeny titers by more than four orders of magnitude, demonstrating the remarkable potency of UbVs as antiviral agents. Our results thereby establish a strategy to produce protein-based inhibitors that could protect against a diverse range of viruses by providing UbVs via mRNA or protein delivery technologies or through transgenic techniques.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/virology , Enzyme Inhibitors/pharmacology , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever, Crimean/virology , Middle East Respiratory Syndrome Coronavirus/drug effects , Ubiquitin/metabolism , Viral Proteins/antagonists & inhibitors , Antiviral Agents/chemistry , Coronavirus Infections/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Hemorrhagic Fever Virus, Crimean-Congo/enzymology , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever, Crimean/metabolism , Humans , Middle East Respiratory Syndrome Coronavirus/enzymology , Middle East Respiratory Syndrome Coronavirus/genetics , Ubiquitination/drug effects , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
5.
J Infect Dis ; 217(12): 1952-1956, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29584885

ABSTRACT

The use of ribavirin to treat Crimean-Congo hemorrhagic fever virus (CCHFV) infection has been controversial, based on uncertainties about its antiviral efficacy in clinical case studies. We studied the effect of ribavirin treatment on viral populations in a recent case by deep-sequencing analysis of plasma samples obtained from a CCHFV-infected patient before, during, and after a 5-day regimen of ribavirin treatment. The CCHFV load dropped during ribavirin treatment, and subclonal diversity (transitions) and indels increased in viral genomes during treatment. Although the results are based on a single case, these data demonstrate the mutagenic effect of ribavirin on CCHFV in vivo.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever, Crimean/drug therapy , Ribavirin/therapeutic use , Antibodies, Viral/immunology , Antiviral Agents/immunology , Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever, Crimean/immunology , Humans
6.
J Assoc Physicians India ; 59: 164-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21751625

ABSTRACT

Crimean Congo hemorrhagic fever (CCHF) has been in the news with reports of its outbreak in India from Gujarat. CCHF is caused by a virus which is a member of the Nairovirus genus of the family Bunyaviridae. All of these viruses are transmitted by either ixodid or argasid ticks. Humans get this infection after a bite of an infected tick or from one infected human to another by contact with infectious blood or body fluids. Workers in livestock and agriculture industry, slaughterhouses, and veterinary practice are most prone to this infection. In severe cases after 3-6 days of the onset of symptoms hemorrhagic manifestations occur. IgG and IgM antibodies may be detected in serum by ELISA from about the sixth day of the illness. The mainstay of treatment in CCHF is supportive. Management of DIC, sepsis, shock and MODS should be undertaken. The antiviral drug Ribavirin has shown benefits. Benefits of treatment with ribavirin outweigh the fatal risks, and ribavirin may therefore be recommended. People at risk should use effective personal protective measures against tick bites. Acaricide treatment of livestock in CCHF virus endemic areas is effective in reducing the population of infected ticks.


Subject(s)
Arachnid Vectors/virology , Hemorrhagic Fever Virus, Crimean-Congo/isolation & purification , Hemorrhagic Fever, Crimean , Ticks/virology , Animals , Antiviral Agents/therapeutic use , Enzyme-Linked Immunosorbent Assay , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever, Crimean/diagnosis , Hemorrhagic Fever, Crimean/drug therapy , Hemorrhagic Fever, Crimean/epidemiology , Hemorrhagic Fever, Crimean/prevention & control , Hemorrhagic Fever, Crimean/transmission , Hemorrhagic Fever, Crimean/virology , Humans , India/epidemiology , Panic , Polymerase Chain Reaction , Prognosis , Ribavirin/therapeutic use , Risk Factors
7.
Viruses ; 13(7)2021 06 22.
Article in English | MEDLINE | ID: mdl-34206476

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a widespread, tick-borne pathogen that causes Crimean-Congo hemorrhagic fever (CCHF) with high morbidity and mortality. CCHFV is transmitted to humans through tick bites or direct contact with patients or infected animals with viremia. Currently, climate change and globalization have increased the transmission risk of this biosafety level (BSL)-4 virus. The treatment options of CCHFV infection remain limited and there is no FDA-approved vaccine or specific antivirals, which urges the identification of potential therapeutic targets and the design of CCHF therapies with greater effort. In this article, we discuss the current progress and some future directions in the development of antiviral strategies against CCHFV.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever, Crimean/drug therapy , Tick-Borne Diseases/drug therapy , Tick-Borne Diseases/virology , Animals , Arachnid Vectors/virology , Hemorrhagic Fever, Crimean/transmission , Hemorrhagic Fever, Crimean/virology , Humans , Mice , Tick-Borne Diseases/transmission , Ticks/virology
8.
Antiviral Res ; 181: 104858, 2020 09.
Article in English | MEDLINE | ID: mdl-32645335

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a widely distributed hemorrhagic fever virus found throughout Eastern Europe, Africa, the Middle East and Asia. It is spread through bites from infected ticks, animal husbandry and can also be acquired in the healthcare setting during care of infected patients. In humans, CCHFV can cause a sudden onset of a non-specific febrile illness that can rapidly progress to severe hemorrhagic manifestations. Currently, there is no widely available vaccine and although ribavirin has been suggested for the treatment of CCHFV, clinical efficacy in both animal models and humans is inconsistent suggesting more potent antivirals are needed for CCHFV. Favipiravir is approved in Japan for the treatment of influenza virus infections and has shown promise against other highly pathogenic RNA viruses including CCHFV with demonstrated efficacy in the type I interferon deficient mouse model. In this report we utilized the cynomolgus macaque model to evaluate the efficacy of once- and twice-daily favipiravir treatment against CCHFV infection. We found that favipiravir treatment suppressed viremia and viral shedding when treatment was initiated 24 h post-infection and viral burdens in key tissues trended lower in favipiravir-treated animals. Our data indicate that favipiravir has efficacy against CCHFV in vivo in a non-human primate model of infection.


Subject(s)
Amides/therapeutic use , Antiviral Agents/therapeutic use , Hemorrhagic Fever, Crimean/drug therapy , Pyrazines/therapeutic use , Viremia/drug therapy , Virus Shedding/drug effects , Animals , Disease Models, Animal , Drug Administration Schedule , Female , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Macaca fascicularis/virology , Male , Viral Load
9.
Viruses ; 12(12)2020 12 10.
Article in English | MEDLINE | ID: mdl-33322045

ABSTRACT

Recent RNA virus outbreaks such as Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Ebola virus (EBOV) have caused worldwide health emergencies highlighting the urgent need for new antiviral strategies. Targeting host cell pathways supporting viral replication is an attractive approach for development of antiviral compounds, especially with new, unexplored viruses where knowledge of virus biology is limited. Here, we present a strategy to identify host-targeted small molecule inhibitors using an image-based phenotypic antiviral screening assay followed by extensive target identification efforts revealing altered cellular pathways upon antiviral compound treatment. The newly discovered antiviral compounds showed broad-range antiviral activity against pathogenic RNA viruses such as SARS-CoV-2, EBOV and Crimean-Congo hemorrhagic fever virus (CCHFV). Target identification of the antiviral compounds by thermal protein profiling revealed major effects on proteostasis pathways and disturbance in interactions between cellular HSP70 complex and viral proteins, illustrating the supportive role of HSP70 on many RNA viruses across virus families. Collectively, this strategy identifies new small molecule inhibitors with broad antiviral activity against pathogenic RNA viruses, but also uncovers novel virus biology urgently needed for design of new antiviral therapies.


Subject(s)
Antiviral Agents/pharmacology , Host-Pathogen Interactions/drug effects , RNA Viruses/drug effects , Virus Replication/drug effects , Animals , Cell Line , Ebolavirus/drug effects , Ebolavirus/physiology , HSP70 Heat-Shock Proteins/metabolism , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Humans , Protein Binding/drug effects , Protein Stability , Proteome/drug effects , Proteostasis/drug effects , RNA Virus Infections/metabolism , RNA Virus Infections/virology , RNA Viruses/physiology , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Small Molecule Libraries/pharmacology , Viral Proteins/metabolism
10.
Jpn J Infect Dis ; 62(1): 11-5, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19168952

ABSTRACT

The mortality rate resulting from Crimean-Congo hemorrhagic fever (CCHF) has been reported in different epidemics to range from 5% to more than 70%. While ribavirin has been recommended as the drug of choice in the treatment of CCHF, no consistent study has unequivocally demonstrated its effectiveness. Using the case-control method, we attempted to evaluate the efficacy of ribavirin in reducing mortality among CCHF cases admitted to Boo-Ali Educational Hospital in Zahedan, Iran, during the years 2000 to 2006. Sixteen deaths among CCHF cases were compared with 47 cases of survival. All patients had a definitive diagnosis based on the results of IgG and IgM capture ELISA tests recorded in their files. Ribavirin therapy for patients who survived had begun on average approximately 24 h earlier than the initiation of ribavirin therapy in the cases of death (P=0.033), and about 2 days earlier in non-bleeding survivors than in bleeding survivors (P=0.013). Based on the results of a multivariable analysis, the most important variables found to enhance survival were the time interval between the disease onset and ribavirin prescription (P=0.004), the time interval between bleeding onset and ribavirin prescription (P=0.037), and the time interval between disease onset and bleeding onset (P=0.014). Based on our findings, ribavirin appears to exert a marked effect on disease outcome, especially when it is prescribed within the first 4 days of the disease.


Subject(s)
Antiviral Agents/therapeutic use , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever, Crimean/drug therapy , Ribavirin/therapeutic use , Administration, Oral , Adolescent , Adult , Aged , Antiviral Agents/pharmacology , Case-Control Studies , Child , Enzyme-Linked Immunosorbent Assay , Female , Hemorrhagic Fever Virus, Crimean-Congo/isolation & purification , Hemorrhagic Fever, Crimean/epidemiology , Humans , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Iran/epidemiology , Male , Middle Aged , Polymerase Chain Reaction , Ribavirin/pharmacology , Survivors , Treatment Outcome
11.
Sci Rep ; 9(1): 6809, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31048746

ABSTRACT

The Crimean-Congo Hemorrhagic Fever virus (CCHFV) is a segmented negative single-stranded RNA virus (-ssRNA) which causes severe hemorrhagic fever in humans with a mortality rate of ~50%. To date, no vaccine has been approved. Treatment is limited to supportive care with few investigational drugs in practice. Previous studies have identified viral RNA dependent RNA Polymerase (RdRp) as a potential drug target due to its significant role in viral replication and transcription. Since no crystal structure is available yet, we report the structural elucidation of CCHFV-RdRp by in-depth homology modeling. Even with low sequence identity, the generated model suggests a similar overall structure as previously reported RdRps. More specifically, the model suggests the presence of structural/functional conserved RdRp motifs for polymerase function, the configuration of uniform spatial arrangement of core RdRp sub-domains, and predicted positively charged entry/exit tunnels, as seen in sNSV polymerases. Extensive pharmacophore modeling based on per-residue energy contribution with investigational drugs allowed the concise mapping of pharmacophoric features and identified potential hits. The combination of pharmacophoric features with interaction energy analysis revealed functionally important residues in the conserved motifs together with in silico predicted common inhibitory binding modes with highly potent reference compounds.


Subject(s)
Antiviral Agents/chemistry , Drug Discovery , Hemorrhagic Fever Virus, Crimean-Congo/enzymology , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Models, Molecular , RNA-Dependent RNA Polymerase/chemistry , Amino Acids , Antiviral Agents/pharmacology , Binding Sites , Catalytic Domain , Drug Discovery/methods , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Humans , Molecular Conformation , Molecular Structure , Protein Binding , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Structure-Activity Relationship , Virus Replication/drug effects
12.
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
13.
Antiviral Res ; 78(1): 125-31, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18096251

ABSTRACT

Crimean-Congo hemorrhagic fever (CCHF) has the most extensive geographic range of the medically significant tick-borne viruses, occurring from western China across southern Asia to eastern Europe and South Africa. The causative agent is a negative-sense, single-stranded RNA virus in the genus Nairovirus, family Bunyaviridae. In published reports, the case fatality rate has generally ranged from 10% to 50%. Sporadic cases and outbreaks of the disease have increased during the past decade across the endemic region. CCHF was first diagnosed in Turkey in 2002, but since then more than 1100 cases have been confirmed by IgM serology or RT-PCR, with a fatality rate of just over 5%. Simple methods are available for the in vitro evaluation of antiviral drugs, but because CCHF virus does not cause disease in its reservoir species or in laboratory animals other than suckling mice, methods are lacking for in vivo efficacy testing. Intravenous or oral ribavirin has been used in several countries to treat the disease for more than 20 years. Evidence of its efficacy is limited to observational studies, and placebo-controlled trials may be impossible to perform for ethical reasons. However, careful analysis of properly stratified observational studies can be used to assess the effects of treatment. This article reviews current approaches to the treatment of CCHF, focusing on the use of ribavirin and hematological support, and discusses prospects for future research.


Subject(s)
Hemorrhagic Fever, Crimean/therapy , Antiviral Agents/therapeutic use , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever Virus, Crimean-Congo/pathogenicity , Hemorrhagic Fever, Crimean/epidemiology , Hemorrhagic Fever, Crimean/physiopathology , Hemorrhagic Fever, Crimean/virology , Humans , Plasma , Platelet Transfusion , Ribavirin/therapeutic use , Turkey/epidemiology
14.
Vopr Virusol ; 53(1): 34-5, 2008.
Article in Russian | MEDLINE | ID: mdl-18318134

ABSTRACT

The high activity of ribavirin made by effective biotechnology in Russia was established in in vitro experiments using the models Crimean-Congo hemorrhagic fever virus, Rift Valley fever virus, and Tahyna and Dhori viruses, which suggests that it is promising in using the drug in the treatment of infection with these viruses.


Subject(s)
Antiviral Agents/pharmacology , Encephalitis Virus, California/drug effects , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Ribavirin/pharmacology , Rift Valley fever virus/drug effects , Thogotovirus/drug effects , Animals , Cell Line, Tumor , Chlorocebus aethiops , Cytopathogenic Effect, Viral/drug effects , Humans , Neutralization Tests , Vero Cells
15.
Antiviral Res ; 157: 18-26, 2018 09.
Article in English | MEDLINE | ID: mdl-29936152

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a cause of serious hemorrhagic disease in humans. Humans infected with CCHFV develop a non-specific febrile illness and then progress to the hemorrhagic phase where case fatality rates can be as high as 30%. Currently there is lack of vaccines and the recommended antiviral treatment, ribavirin, has inconsistent efficacy in both human and animal studies. In this study we developed a model of CCHFV infection in type I interferon deficient mice using the clinical CCHFV isolate strain Hoti. Mice infected with strain Hoti develop a progressively worsening and ultimately fatal disease. We utilized this model along with our established model using the prototypical CCHFV strain 10200 to evaluate treatment with ribavirin or the antiviral favipiravir. While ribavirin treatment was able to suppress viral loads at early time points it was ultimately unable to prevent development of terminal disease in mice infected with either strain of CCHFV. In contrast, favipiravir showed clinical benefit even when administered late in the clinical progression of CCHF. Interestingly, in a small subset of mice, late-onset of CCHF was observed after favipiravir treatment was stopped and persistence of viral RNA in favipiravir treated survivors was also seen. Nevertheless, favipiravir showed significant clinical benefit against two distinct strains of CCHFV suggesting it may be a potent antiviral for treatment of human CCHFV infections.


Subject(s)
Amides/administration & dosage , Antiviral Agents/administration & dosage , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever, Crimean/drug therapy , Pyrazines/administration & dosage , Ribavirin/administration & dosage , Amides/pharmacology , Animals , Antiviral Agents/pharmacology , Disease Models, Animal , Hemorrhagic Fever Virus, Crimean-Congo/growth & development , Hemorrhagic Fever, Crimean/pathology , Mice , Pyrazines/pharmacology , RNA, Viral/analysis , Ribavirin/pharmacology , Survival Analysis , Treatment Outcome , Viral Load
16.
Comput Biol Chem ; 70: 1-6, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28709136

ABSTRACT

Crimean-Congo Hemorrhagic Fever Virus (CCHFV) is one of the deadliest human diseases with mortality rate near 50%. Special attention should be paid to this virus since there is no approved treatment for it. On the other hand, the recent outbreak of Ebola virus which is a member of hemorrhagic fever viruses shows this group of viruses can be extremely dangerous. Previous studies have indicated that nucleoprotein of CCHFV, a pivotal protein in virus replication, is an appropriate target for antiviral drug development. The aim of this study is finding inhibitor(s) of this protein. Herein, a virtual screening procedure employing docking followed by molecular dynamic was used to identify small molecule inhibitors of the nucleoprotein from FDA-approved drugs. Regarding CCHFV, using in-silico method is a safe way to achieve its inhibitor(s) since this virus is categorized as a World Health Organization (WHO) biosafety level 4 pathogen and therefore investigation in general laboratories is restricted. In conclusion, considering docking and molecular dynamic results alongside with bioavailability of FDA-approved drugs, doxycycline and minocycline are proposed as potential inhibitors of CCHFV nucleoprotein. There is hope, this study encourage other research groups for in-vitro and in-vivo studies about the efficacy of those two medicines in CCHFV treatment.


Subject(s)
Antiviral Agents/pharmacology , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Molecular Docking Simulation , Molecular Dynamics Simulation , Tetracyclines/pharmacology , Antiviral Agents/chemistry , Microbial Sensitivity Tests , Tetracyclines/chemistry
17.
Antiviral Res ; 147: 91-99, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29024765

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV), a tick-borne orthonairovirus, causes a severe hemorrhagic disease in humans (Crimean-Congo hemorrhagic fever, CCHF). Currently, no vaccines are approved to prevent CCHF; treatment is limited to supportive care and the use of ribavirin, the therapeutic benefits of which remain unclear. CCHF is part of WHO's priority list of infectious diseases warranting further research and development. To aid in the identification of new antiviral compounds, we generated a recombinant CCHFV expressing a reporter protein, allowing us to quantify virus inhibition by measuring the reduction in fluorescence in infected cells treated with candidate compounds. The screening assay was readily adaptable to high-throughput screening (HTS) of compounds using Huh7 cells, with a signal-to-noise ratio of 50:1, and Z'-factors > 0.6 in both 96- and 384-well formats. A screen of candidate nucleoside analog compounds identified 2'-deoxy-2'-fluorocytidine (EC50 = 61 ± 18 nM) as having 200 × the potency of ribavirin (EC50 = 12.5 ± 2.6 µM), as well as 17 × the potency of T-705 (favipiravir), another compound with reported anti-CCHFV activity (EC50 = 1.03 ± 0.16 µM). Furthermore, we also determined that 2'-deoxy-2'-fluorocytidine acts synergistically with T-705 to inhibit CCHFV replication without causing cytotoxicity. The incorporation of this reporter virus into the high-throughput screening assay described here will allow more rapid identification of effective therapeutic options to combat this emerging human pathogen.


Subject(s)
Deoxycytidine/analogs & derivatives , Drug Discovery/methods , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , High-Throughput Screening Assays , Virus Replication/drug effects , Amides/pharmacology , Antiviral Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Genes, Reporter/genetics , Green Fluorescent Proteins/genetics , Hemorrhagic Fever, Crimean/virology , Humans , Lethal Dose 50 , Pyrazines/pharmacology , Ribavirin/pharmacology , Viral Proteins/genetics
18.
Virus Res ; 120(1-2): 184-90, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16632039

ABSTRACT

Crimean Congo hemorrhagic fever virus (CCHFV) is a geographically widespread pathogen that causes severe hemorrhagic fever with high mortality. Even though one of the main objectives focuses on the progress of antiviral agents, the research on CCHFV is strongly hampered due to its BSL-4 classification. Nitric oxide (NO), a mediator with broad biological effects, has been shown to possess inhibitory properties against various pathogens. The molecule constitutes a component of the innate immunity and serves to assist in the early immunological events where it contributes to clearance of microorganisms. In this study, we investigated the inhibitory properties of exogenous NO on CCHFV. We found that NO had a significant antiviral activity against CCHFV replication. By using the NO-donor S-nitroso-N-acetylpenicillamine (SNAP) we were able to show up to 99% reduction in virion progeny yield. In contrast, 3-morpholinosydnonimine hydrochloride (SIN-1), a peroxynitrite donor, had no significant antiviral activity against CCHFV. Furthermore the expression of viral proteins; the nucleocapsid protein and the glycoprotein, were clearly reduced with increasing concentrations of SNAP. We have also shown that the amount of total vRNA in SNAP-treated cells was reduced by about 50% compared to the controls.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Nitric Oxide Donors/pharmacology , S-Nitroso-N-Acetylpenicillamine/pharmacology , Dose-Response Relationship, Drug , Glycoproteins/metabolism , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Nitric Oxide Donors/metabolism , Nucleocapsid Proteins/metabolism , RNA, Viral/biosynthesis , S-Nitroso-N-Acetylpenicillamine/metabolism , Virus Replication
19.
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
20.
Antiviral Res ; 118: 75-81, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25796972

ABSTRACT

Crimean-Congo hemorrhagic virus (CCHFV) causes hemorrhagic fever with high case mortality rates and is endemic in south-eastern Europe, Africa, and Asia. The limited catalog of specific treatment, highlight the necessity to look for additional therapeutic solutions. Previous experiments suggested that CCHFV enters the cells via a clathrin dependent pathway. Therefore, we have evaluated the potential anti-CCHFV activity of several molecules targeting this entry possibility. We identified two molecules chloroquine and chlorpromazine. Neutralization and virus yield reduction assays were tested in Vero E6 and Huh7 cells on two different CCHFV strains. Several combinations, including ribavirin, were assayed to test a potential synergistic effect. The two molecules inhibited CCHFV, and depending on the virus and the cell lines, the 50% inhibitory concentration (IC50) values for chloroquine and chlorpromazine ranged from 28 to 43 and 10.8-15.7 µM, respectively. Time-of-addition studies demonstrated that these molecules had a direct effect on CCHFV infectivity and spread. The antiviral activity of the two molecules was still effective even when added up to 6h post-infection and up to 24h. The selectivity index ranging from 3 to 35 lead us to evaluate combinations with ribavirin. Combinations of ribavirin and chloroquine or chlorpromazine were synergistic against CCHFV. Though the low chlorpromazine selectivity index suggests the need for a chemical improvement, our present study highlights chloroquine as the main drug having the potential for drug repurposing.


Subject(s)
Antiviral Agents/pharmacology , Chloroquine/pharmacology , Chlorpromazine/pharmacology , Drug Repositioning , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Virus Internalization/drug effects , Animals , Cell Line , Drug Synergism , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Humans , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Ribavirin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL