51
|
Fox JM, Diamond MS. Immune-Mediated Protection and Pathogenesis of Chikungunya Virus. THE JOURNAL OF IMMUNOLOGY 2017; 197:4210-4218. [PMID: 27864552 DOI: 10.4049/jimmunol.1601426] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 09/13/2016] [Indexed: 01/16/2023]
Abstract
Chikungunya virus (CHIKV) is a re-emerging alphavirus that causes debilitating acute and chronic arthritis. Infection by CHIKV induces a robust immune response that is characterized by production of type I IFNs, recruitment of innate and adaptive immune cells, and development of neutralizing Abs. Despite this response, chronic arthritis can develop in some individuals, which may be due to a failure to eliminate viral RNA and Ag and/or persistent immune responses that cause chronic joint inflammation. In this review, based primarily on advances from recent studies in mice, we discuss the innate and adaptive immune factors that control CHIKV dissemination and clearance or contribute to pathogenesis.
Collapse
Affiliation(s)
- Julie M Fox
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110; .,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110; and.,Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
52
|
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus in the family Togaviridae that causes outbreaks of debilitating acute and chronic arthralgia in humans. Although historically associated with localized outbreaks in Africa and Asia, recent epidemics in the Indian Ocean region and the Americas have led to the recognition that CHIKV is capable of moving into previously unaffected areas and causing significant levels of human suffering. The severity of CHIKV rheumatic disease, which can severely impact life quality of infected individuals for weeks, months, or even years, combined with the explosive nature of CHIKV outbreaks and its demonstrated ability to quickly spread into new regions, has led to renewed interest in developing strategies for the prevention or treatment of CHIKV-induced disease. Therefore, this chapter briefly discusses the biology of CHIKV and the factors contributing to CHIKV dissemination, while also discussing the pathogenesis of CHIKV-induced disease and summarizing the status of efforts to develop safe and effective therapies and vaccines against CHIKV and related viruses.
Collapse
|
53
|
Development of a Novel Virus-Like Particle Vaccine Platform That Mimics the Immature Form of Alphavirus. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00090-17. [PMID: 28515133 PMCID: PMC5498722 DOI: 10.1128/cvi.00090-17] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/04/2017] [Indexed: 12/27/2022]
Abstract
Virus-like particles (VLPs) are noninfectious multiprotein structures that are engineered to self-assemble from viral structural proteins. Here, we developed a novel VLP-based vaccine platform utilizing VLPs from the chikungunya virus. We identified two regions within the envelope protein, a structural component of chikungunya, where foreign antigens can be inserted without compromising VLP structure. Our VLP displays 480 copious copies of an inserted antigen on the VLP surface in a highly symmetric manner and is thus capable of inducing strong immune responses against any inserted antigen. Furthermore, by mimicking the structure of the immature form of the virus, we altered our VLP's in vivo dynamics and enhanced its immunogenicity. We used the circumsporozoite protein (CSP) of the Plasmodium falciparum malaria parasite as an antigen and demonstrated that our VLP-based vaccine elicits strong immune responses against CSP in animals. The sera from immunized monkeys protected mice from malaria infection. Likewise, mice vaccinated with P. yoelii CSP-containing VLPs were protected from an infectious sporozoite challenge. Hence, our uniquely engineered VLP platform can serve as a blueprint for the development of vaccines against other pathogens and diseases.
Collapse
|
54
|
Abstract
Chikungunya virus (CHIKV) is an arthropod-borne alphavirus that causes acute and chronic arthritis. The virus reemerged in the Indian Ocean islands in 2005-2006 and is responsible for outbreaks in the Caribbean islands and the Americas since late 2013. Despite the wealth of research over the past 10 years, there are no commercially available antiviral drugs or vaccines. Treatment usually involves analgesics, anti-inflammatory drugs, and supportive care. Most studies have been focused on understanding the pathogenesis of CHIKV infection through clinical observation and with animal models. In this review, the clinical manifestations of CHIKV that define the disease and the use of relevant animal models, from mice to nonhuman primates, are discussed. Understanding key cellular factors in CHIKV infection and the interplay with the immune system will aid in the development of preventive and therapeutic approaches to combat this painful viral disease in humans.
Collapse
Affiliation(s)
- Lisa F P Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648; .,Institute of Infection and Global Health, University of Liverpool, Liverpool L69 3BX, United Kingdom
| |
Collapse
|
55
|
Therapeutic administration of a recombinant human monoclonal antibody reduces the severity of chikungunya virus disease in rhesus macaques. PLoS Negl Trop Dis 2017. [PMID: 28628616 PMCID: PMC5491320 DOI: 10.1371/journal.pntd.0005637] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne virus that causes a febrile syndrome in humans associated with acute and chronic debilitating joint and muscle pain. Currently no licensed vaccines or therapeutics are available to prevent or treat CHIKV infections. We recently isolated a panel of potently neutralizing human monoclonal antibodies (mAbs), one (4N12) of which exhibited prophylactic and post-exposure therapeutic activity against CHIKV in immunocompromised mice. Here, we describe the development of an engineered CHIKV mAb, designated SVIR001, that has similar antigen binding and neutralization profiles to its parent, 4N12. Because therapeutic administration of SVIR001 in immunocompetent mice significantly reduced viral load in joint tissues, we evaluated its efficacy in a rhesus macaque model of CHIKV infection. Rhesus macaques that were treated after infection with SVIR001 showed rapid elimination of viremia and less severe joint infiltration and disease compared to animals treated with SVIR002, an isotype control mAb. SVIR001 reduced viral burden at the site of infection and at distant sites and also diminished the numbers of activated innate immune cells and levels of pro-inflammatory cytokines and chemokines. SVIR001 therapy; however, did not substantively reduce the induction of CHIKV-specific B or T cell responses. Collectively, these results show promising therapeutic activity of a human anti-CHIKV mAb in rhesus macaques and provide proof-of-principle for its possible use in humans to treat active CHIKV infections. Chikungunya virus (CHIKV) causes fever, rash, and acute and chronic arthralgia. Currently there are no approved therapies to treat or vaccines to prevent CHIKV infection in humans. In this study, we engineered SVIR001, a recombinant fully human monoclonal antibody (mAb) that eliminated viremia, reduced viral load at the site of infection, and diminished spread to distant target tissues in rhesus macaques when administered after infection. SVIR001 treatment reduced joint inflammation and disease without impairing the induction of the adaptive immune response. These results demonstrate the efficacy of mAb therapy to reduce the severity of CHIKV disease.
Collapse
|
56
|
Clayton AM. Monoclonal Antibodies as Prophylactic and Therapeutic Agents Against Chikungunya Virus. J Infect Dis 2017; 214:S506-S509. [PMID: 27920182 DOI: 10.1093/infdis/jiw324] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that is responsible for considerable epidemics worldwide and recently emerged in the Americas in 2013. CHIKV may cause long-lasting arthralgia after acute infection. With currently no licensed vaccines or antivirals, the design of effective therapies to prevent or treat CHIKV infection is of utmost importance and will be facilitated by increased understanding of the dynamics of chikungunya. In this article, monoclonal antibodies against CHIKV as viable prophylactic and therapeutic agents will be discussed.
Collapse
Affiliation(s)
- April M Clayton
- Office of Global Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
57
|
Affiliation(s)
- David M Vu
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, G312, Stanford, CA 94305, USA.
| | - Donald Jungkind
- St. George's University School of Medicine, Grenada, West Indies
| | - Angelle Desiree LaBeaud
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, G312, Stanford, CA 94305, USA
| |
Collapse
|
58
|
Roques P, Ljungberg K, Kümmerer BM, Gosse L, Dereuddre-Bosquet N, Tchitchek N, Hallengärd D, García-Arriaza J, Meinke A, Esteban M, Merits A, Le Grand R, Liljeström P. Attenuated and vectored vaccines protect nonhuman primates against Chikungunya virus. JCI Insight 2017; 2:e83527. [PMID: 28352649 DOI: 10.1172/jci.insight.83527] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chikungunya virus (CHIKV) is rapidly spreading across the globe, and millions are infected. Morbidity due to this virus is a serious threat to public health, but at present, there is no vaccine against this debilitating disease. We have recently developed a number of vaccine candidates, and here we have evaluated 3 of them in a nonhuman primate model. A single immunization with an attenuated strain of CHIKV (Δ5nsP3), a homologous prime-boost immunization with a DNA-launched RNA replicon encoding CHIKV envelope proteins (DREP-E), and a DREP-E prime followed by a recombinant modified vaccinia virus Ankara encoding CHIKV capsid and envelope (MVA-CE) boost all induced protection against WT CHIKV infection. The attenuated Δ5nsP3 virus proved to be safe and did not show any clinical signs typically associated with WT CHIKV infections such as fever, skin rash, lymphopenia, or joint swelling. These vaccines are based on an East/Central/South African strain of Indian Ocean lineage, but they also generated neutralizing antibodies against an isolate of the Asian genotype that now is rapidly spreading across the Americas. These results form the basis for clinical development of an efficacious CHIKV vaccine that generates both humoral and cellular immunity with long-term immunological memory.
Collapse
Affiliation(s)
- Pierre Roques
- Université Paris Sud, UMR 1184, Orsay, France.,CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT center.,Inserm, U1184, Center for immunology of viral infections and autoimmune diseases, Fontenay aux Roses, France
| | - Karl Ljungberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Beate M Kümmerer
- Institute of Virology, University of Bonn Medical Centre, Bonn, Germany
| | - Leslie Gosse
- Université Paris Sud, UMR 1184, Orsay, France.,CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT center.,Inserm, U1184, Center for immunology of viral infections and autoimmune diseases, Fontenay aux Roses, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris Sud, UMR 1184, Orsay, France.,CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT center.,Inserm, U1184, Center for immunology of viral infections and autoimmune diseases, Fontenay aux Roses, France
| | - Nicolas Tchitchek
- Université Paris Sud, UMR 1184, Orsay, France.,CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT center.,Inserm, U1184, Center for immunology of viral infections and autoimmune diseases, Fontenay aux Roses, France
| | - David Hallengärd
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | | | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Roger Le Grand
- Université Paris Sud, UMR 1184, Orsay, France.,CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT center.,Inserm, U1184, Center for immunology of viral infections and autoimmune diseases, Fontenay aux Roses, France
| | - Peter Liljeström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
59
|
Silva LA, Dermody TS. Chikungunya virus: epidemiology, replication, disease mechanisms, and prospective intervention strategies. J Clin Invest 2017; 127:737-749. [PMID: 28248203 PMCID: PMC5330729 DOI: 10.1172/jci84417] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chikungunya virus (CHIKV), a reemerging arbovirus, causes a crippling musculoskeletal inflammatory disease in humans characterized by fever, polyarthralgia, myalgia, rash, and headache. CHIKV is transmitted by Aedes species of mosquitoes and is capable of an epidemic, urban transmission cycle with high rates of infection. Since 2004, CHIKV has spread to new areas, causing disease on a global scale, and the potential for CHIKV epidemics remains high. Although CHIKV has caused millions of cases of disease and significant economic burden in affected areas, no licensed vaccines or antiviral therapies are available. In this Review, we describe CHIKV epidemiology, replication cycle, pathogenesis and host immune responses, and prospects for effective vaccines and highlight important questions for future research.
Collapse
|
60
|
RNA-Seq analysis of chikungunya virus infection and identification of granzyme A as a major promoter of arthritic inflammation. PLoS Pathog 2017; 13:e1006155. [PMID: 28207896 PMCID: PMC5312928 DOI: 10.1371/journal.ppat.1006155] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 12/28/2016] [Indexed: 02/07/2023] Open
Abstract
Chikungunya virus (CHIKV) is an arthritogenic alphavirus causing epidemics of acute and chronic arthritic disease. Herein we describe a comprehensive RNA-Seq analysis of feet and lymph nodes at peak viraemia (day 2 post infection), acute arthritis (day 7) and chronic disease (day 30) in the CHIKV adult wild-type mouse model. Genes previously shown to be up-regulated in CHIKV patients were also up-regulated in the mouse model. CHIKV sequence information was also obtained with up to ≈8% of the reads mapping to the viral genome; however, no adaptive viral genome changes were apparent. Although day 2, 7 and 30 represent distinct stages of infection and disease, there was a pronounced overlap in up-regulated host genes and pathways. Type I interferon response genes (IRGs) represented up to ≈50% of up-regulated genes, even after loss of type I interferon induction on days 7 and 30. Bioinformatic analyses suggested a number of interferon response factors were primarily responsible for maintaining type I IRG induction. A group of genes prominent in the RNA-Seq analysis and hitherto unexplored in viral arthropathies were granzymes A, B and K. Granzyme A-/- and to a lesser extent granzyme K-/-, but not granzyme B-/-, mice showed a pronounced reduction in foot swelling and arthritis, with analysis of granzyme A-/- mice showing no reductions in viral loads but reduced NK and T cell infiltrates post CHIKV infection. Treatment with Serpinb6b, a granzyme A inhibitor, also reduced arthritic inflammation in wild-type mice. In non-human primates circulating granzyme A levels were elevated after CHIKV infection, with the increase correlating with viral load. Elevated granzyme A levels were also seen in a small cohort of human CHIKV patients. Taken together these results suggest granzyme A is an important driver of arthritic inflammation and a potential target for therapy. Trial Registration: ClinicalTrials.gov NCT00281294 The largest chikungunya virus (CHIKV) epidemic ever recorded began in 2004 in Africa and spread across Asia reaching Europe and recently the Americas, with millions of cases reported. We undertook a detailed analysis of the mRNA expression profile during acute and chronic arthritis in an adult wild-type mouse model of CHIKV infection and disease. Gene induction profiles showed a high concordance with published human data, providing some validation of the mouse model. The host response was overwhelmingly dominated by type I interferon response genes, even after type I interferon induction was lost. The analysis also provided information on CHIKV RNA, with no adaptive viral genome changes identified. An important goal of the analysis was to identify new players in arthritic inflammation. Granzyme A was prominent in the RNA-Seq data and granzyme A deficient mice showed reduced arthritis, with no effects on viral loads. Arthritic disease could also be ameliorated in wild-type mice with a granzyme A inhibitor. Elevated circulating granzyme A levels were seen in non-human primates infected with CHIKV and in human CHIKV patients. Granzyme A thus emerges to be a major driver of CHIKV-mediated arthritic inflammation and a potential target for anti-inflammatory interventions.
Collapse
|
61
|
The neutralizing role of IgM during early Chikungunya virus infection. PLoS One 2017; 12:e0171989. [PMID: 28182795 PMCID: PMC5300252 DOI: 10.1371/journal.pone.0171989] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/30/2017] [Indexed: 12/13/2022] Open
Abstract
The antibody isotype IgM appears earlier than IgG, within days of onset of symptoms, and is important during the early stages of the adaptive immune response. Little is known about the functional role of IgM during infection with chikungunya virus (CHIKV), a recently reemerging arbovirus that has caused large global outbreaks. In this study, we studied antibody responses in 102 serum samples collected during CHIKV outbreaks in Malaysia. We described the neutralizing role of IgM at different times post-infection and examined the independent contributions of IgM and IgG towards the neutralizing capacity of human immune sera during the early phase of infection, including the differences in targets of neutralizing epitopes. Neutralizing IgM starts to appear as early as day 4 of symptoms, and their appearance from day 6 is associated with a reduction in viremia. IgM acts in a complementary manner with the early IgG, but plays the main neutralizing role up to a point between days 4 and 10 which varies between individuals. After this point, total neutralizing capacity is attributable almost entirely to the robust neutralizing IgG response. IgM preferentially binds and targets epitopes on the CHIKV surface E1-E2 glycoproteins, rather than individual E1 or E2. These findings provide insight into the early antibody responses to CHIKV, and have implications for design of diagnostic serological assays.
Collapse
|
62
|
Burt FJ, Chen W, Miner JJ, Lenschow DJ, Merits A, Schnettler E, Kohl A, Rudd PA, Taylor A, Herrero LJ, Zaid A, Ng LFP, Mahalingam S. Chikungunya virus: an update on the biology and pathogenesis of this emerging pathogen. THE LANCET. INFECTIOUS DISEASES 2017; 17:e107-e117. [PMID: 28159534 DOI: 10.1016/s1473-3099(16)30385-1] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 08/26/2016] [Accepted: 09/23/2016] [Indexed: 12/14/2022]
Abstract
Re-emergence of chikungunya virus, a mosquito-transmitted pathogen, is of serious public health concern. In the past 15 years, after decades of infrequent, sporadic outbreaks, the virus has caused major epidemic outbreaks in Africa, Asia, the Indian Ocean, and more recently the Caribbean and the Americas. Chikungunya virus is mainly transmitted by Aedes aegypti mosquitoes in tropical and subtropical regions, but the potential exists for further spread because of genetic adaptation of the virus to Aedes albopictus, a species that thrives in temperate regions. Chikungunya virus represents a substantial health burden to affected populations, with symptoms that include severe joint and muscle pain, rashes, and fever, as well as prolonged periods of disability in some patients. The inflammatory response coincides with raised levels of immune mediators and infiltration of immune cells into infected joints and surrounding tissues. Animal models have provided insights into disease pathology and immune responses. Although host innate and adaptive responses have a role in viral clearance and protection, they can also contribute to virus-induced immune pathology. Understanding the mechanisms of host immune responses is essential for the development of treatments and vaccines. Inhibitory compounds targeting key inflammatory pathways, as well as attenuated virus vaccines, have shown some success in animal models, including an attenuated vaccine strain based on an isolate from La Reunion incorporating an internal ribosome entry sequence that prevents the virus from infecting mosquitoes and a vaccine based on virus-like particles expressing envelope proteins. However, immune correlates of protection, as well as the safety of prophylactic and therapeutic candidates, are important to consider for their application in chikungunya infections. In this Review, we provide an update on chikungunya virus with regard to its epidemiology, molecular virology, virus-host interactions, immunological responses, animal models, and potential antiviral therapies and vaccines.
Collapse
Affiliation(s)
- Felicity J Burt
- National Health Laboratory Services, Universitas and Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa.
| | - Weiqiang Chen
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Jonathan J Miner
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Deborah J Lenschow
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | | | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Penny A Rudd
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Adam Taylor
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Lara J Herrero
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Ali Zaid
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Lisa F P Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore; Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Suresh Mahalingam
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
63
|
Miner JJ, Cook LE, Hong JP, Smith AM, Richner JM, Shimak RM, Young AR, Monte K, Poddar S, Crowe JE, Lenschow DJ, Diamond MS. Therapy with CTLA4-Ig and an antiviral monoclonal antibody controls chikungunya virus arthritis. Sci Transl Med 2017; 9:eaah3438. [PMID: 28148840 PMCID: PMC5448557 DOI: 10.1126/scitranslmed.aah3438] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/21/2016] [Accepted: 12/02/2016] [Indexed: 12/12/2022]
Abstract
In 2013, chikungunya virus (CHIKV) transmission was documented in the Western Hemisphere, and the virus has since spread throughout the Americas with more than 1.8 million people infected in more than 40 countries. CHIKV targets the joints, resulting in symmetric polyarthritis that clinically mimics rheumatoid arthritis and can endure for months to years. At present, no approved treatment is effective in preventing or controlling CHIKV infection or disease. We treated mice with eight different disease-modifying antirheumatic drugs and identified CLTA4-Ig (abatacept) and tofacitinib as candidate therapies based on their ability to decrease acute joint swelling. CTLA4-Ig reduced T cell accumulation in the joints of infected animals without affecting viral infection. Whereas monotherapy with CTLA4-Ig or a neutralizing anti-CHIKV human monoclonal antibody provided partial clinical improvement, therapy with both abolished swelling and markedly reduced levels of chemokines, proinflammatory cytokines, and infiltrating leukocytes. Thus, combination CTLA4-Ig and antiviral antibody therapy controls acute CHIKV infection and arthritis and may be a candidate for testing in humans.
Collapse
Affiliation(s)
- Jonathan J Miner
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lindsey E Cook
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jun P Hong
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amber M Smith
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Justin M Richner
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Raeann M Shimak
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alissa R Young
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kristen Monte
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Subhajit Poddar
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James E Crowe
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Deborah J Lenschow
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
64
|
Deng CL, Liu SQ, Zhou DG, Xu LL, Li XD, Zhang PT, Li PH, Ye HQ, Wei HP, Yuan ZM, Qin CF, Zhang B. Development of Neutralization Assay Using an eGFP Chikungunya Virus. Viruses 2016; 8:v8070181. [PMID: 27367716 PMCID: PMC4974516 DOI: 10.3390/v8070181] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/31/2016] [Accepted: 06/23/2016] [Indexed: 12/24/2022] Open
Abstract
Chikungunya virus (CHIKV), a member of the Alphavirus genus, is an important human emerging/re-emerging pathogen. Currently, there are no effective antiviral drugs or vaccines against CHIKV infection. Herein, we construct an infectious clone of CHIKV and an eGFP reporter CHIKV (eGFP-CHIKV) with an isolated strain (assigned to Asian lineage) from CHIKV-infected patients. The eGFP-CHIKV reporter virus allows for direct visualization of viral replication through the levels of eGFP expression. Using a known CHIKV inhibitor, ribavirin, we confirmed that the eGFP-CHIKV reporter virus could be used to identify inhibitors against CHIKV. Importantly, we developed a novel and reliable eGFP-CHIKV reporter virus-based neutralization assay that could be used for rapid screening neutralizing antibodies against CHIKV.
Collapse
Affiliation(s)
- Cheng-Lin Deng
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Si-Qing Liu
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Dong-Gen Zhou
- Ningbo International Travel Healthcare Center, Ningbo 315012, China.
| | - Lin-Lin Xu
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Xiao-Dan Li
- School of Medicine, Hunan Normal University, Changsha 410000, China.
| | - Pan-Tao Zhang
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Peng-Hui Li
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Han-Qing Ye
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Hong-Ping Wei
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Zhi-Ming Yuan
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| |
Collapse
|
65
|
Adouchief S, Smura T, Vapalahti O, Hepojoki J. Mapping of human B-cell epitopes of Sindbis virus. J Gen Virol 2016; 97:2243-2254. [PMID: 27339177 DOI: 10.1099/jgv.0.000531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mosquito-transmitted Sindbis virus (SINV) causes fever, skin lesions and musculoskeletal symptoms if transmitted to man. SINV is the prototype virus of genus Alphavirus, which includes other arthritogenic viruses such as chikungunya virus (CHIKV) and Ross River virus (RRV) that cause large epidemics with a considerable public health burden. Until now the human B-cell epitopes have been studied for CHIKV and RRV, but not for SINV. To identify the B-cell epitopes in SINV-infection, we synthetised a library of linear 18-mer peptides covering the structural polyprotein of SINV, and probed it with SINV IgG-positive and IgG-negative serum pools. By comparing the binding profiles of the pools, we identified 15 peptides that were strongly reactive only with the SINV IgG-positive pools. We then utilized alanine scanning and individual (n=22) patient sera to further narrow the number of common B-cell epitopes to six. These epitopes locate to the capsid, E2, E1 and to a region in PE2 (uncleaved E3-E2), which may only be present in immature virions. By sequence comparison, we observed that one of the capsid protein epitopes shares six identical amino acids with macrophage migration inhibitory factor (MIF) receptor, which is linked to inflammatory diseases and to molecular pathology of alphaviral arthritides. Our results add to the current understanding on SINV disease and raise questions of a potential role of uncleaved PE2 and the MIF receptor (CD74) mimotope in human SINV infection.
Collapse
Affiliation(s)
- Samuel Adouchief
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Teemu Smura
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland.,Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland.,Department of Virology and Immunology, Hospital District of Helsinki and Uusimaa (HUSLAB), Helsinki, Finland
| | - Jussi Hepojoki
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
66
|
Ahola T, Couderc T, Courderc T, Ng LFP, Hallengärd D, Powers A, Lecuit M, Esteban M, Merits A, Roques P, Liljeström P. Therapeutics and vaccines against chikungunya virus. Vector Borne Zoonotic Dis 2016; 15:250-7. [PMID: 25897811 DOI: 10.1089/vbz.2014.1681] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Currently, there are no licensed vaccines or therapies available against chikungunya virus (CHIKV), and these were subjects discussed during a CHIKV meeting recently organized in Langkawi, Malaysia. In this review, we chart the approaches taken in both areas. Because of a sharp increase in new data in these fields, the present paper is complementary to previous reviews by Weaver et al. in 2012 and Kaur and Chu in 2013 . The most promising antivirals so far discovered are reviewed, with a special focus on the virus-encoded replication proteins as potential targets. Within the vaccines in development, our review emphasizes the various strategies in parallel development that are unique in the vaccine field against a single disease.
Collapse
Affiliation(s)
- Tero Ahola
- 1 Department of Food and Environmental Sciences, University of Helsinki , Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Characterization of N-glycosylation profiles from mammalian and insect cell derived chikungunya VLP. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1032:218-223. [PMID: 27157808 DOI: 10.1016/j.jchromb.2016.04.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/04/2016] [Accepted: 04/11/2016] [Indexed: 11/21/2022]
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that causes severe arthralgia. The envelope of CHIKV is composed of 240 copies of two glycoproteins: E1 and E2. In this work, we have characterized the N-glycosylation patterns of CHIKV virus-like particles (VLPs), containing both E1 and E2 proteins, derived from mammalian and insect cells using hydrophilic interaction liquid chromatography (HILIC) with fluorescence (FL) and mass spectrometry (MS) detection. While HEK293 derived CHIKV VLPs contain oligomannose, hybrid and complex glycans, VLPs derived from SfBasic predominantly contain oligomannose glycans. This strong host dependence of N-glycosylation pattern resembles other alphaviruses such as SINV. The VLPs from HEK293 and SfBasic, with significantly different N-glycosylation profiles, are valuable reagents enabling future in-depth correlation studies between immunogenicity and glycosylation. In addition, the characterization tools presented here allow one to monitor glycosylation during vaccine process development and ensure process consistency.
Collapse
|
68
|
Roosenhoff R, Anfasa F, Martina B. The pathogenesis of chronic chikungunya: evolving concepts. Future Virol 2016. [DOI: 10.2217/fvl.15.107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Chikungunya virus (CHIKV) re-emerged and caused an outbreak in the Caribbean and the Americas. CHIKV can cause incapacitating arthralgia, which may be evolved in chronic arthritis that is similar to rheumatoid arthritis that lasts for months or years. This review provides an overview of known and hypothesized mechanisms that CHIKV uses to promote chronic arthritis. We hypothesized that the chronic inflammatory response that is stimulated by persisting CHIKV replication in the joints results in the arthritic symptoms seen in patients. Most hypotheses proposed in this review need to be tested or confirmed, which may help in the development of new specific treatments and vaccines against CHIKV that will not only combat viral persistence but also prevent tissue damage.
Collapse
Affiliation(s)
- Rueshandra Roosenhoff
- ARTEMIS One Health Research Institute, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
- Curacao Biomedical & Health Research Institute, Curacao
| | - Fatih Anfasa
- Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Byron Martina
- ARTEMIS One Health Research Institute, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
69
|
Jin J, Liss NM, Chen DH, Liao M, Fox JM, Shimak RM, Fong RH, Chafets D, Bakkour S, Keating S, Fomin ME, Muench MO, Sherman MB, Doranz BJ, Diamond MS, Simmons G. Neutralizing Monoclonal Antibodies Block Chikungunya Virus Entry and Release by Targeting an Epitope Critical to Viral Pathogenesis. Cell Rep 2015; 13:2553-2564. [PMID: 26686638 PMCID: PMC4720387 DOI: 10.1016/j.celrep.2015.11.043] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/15/2015] [Accepted: 11/11/2015] [Indexed: 11/26/2022] Open
Abstract
We evaluated the mechanism by which neutralizing human monoclonal antibodies inhibit chikungunya virus (CHIKV) infection. Potently neutralizing antibodies (NAbs) blocked infection at multiple steps of the virus life cycle, including entry and release. Cryo-electron microscopy structures of Fab fragments of two human NAbs and chikungunya virus-like particles showed a binding footprint that spanned independent domains on neighboring E2 subunits within one viral spike, suggesting a mechanism for inhibiting low-pH-dependent membrane fusion. Detailed epitope mapping identified amino acid E2-W64 as a critical interaction residue. An escape mutation (E2-W64G) at this residue rendered CHIKV attenuated in mice. Consistent with these data, CHIKV-E2-W64G failed to emerge in vivo under the selection pressure of one of the NAbs, IM-CKV063. As our study suggests that antibodies engaging the residue E2-W64 can potently inhibit CHIKV at multiple stages of infection, antibody-based therapies or immunogens that target this region might have protective value.
Collapse
Affiliation(s)
- Jing Jin
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nathan M Liss
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dong-Hua Chen
- Department of Structural Biology, Stanford University, Stanford, CA 94305, USA
| | - Maofu Liao
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Julie M Fox
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Raeann M Shimak
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachel H Fong
- Integral Molecular Inc., Philadelphia, PA 19104, USA
| | - Daniel Chafets
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sonia Bakkour
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sheila Keating
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marina E Fomin
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marcus O Muench
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael B Sherman
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Graham Simmons
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
70
|
Long F, Fong RH, Austin SK, Chen Z, Klose T, Fokine A, Liu Y, Porta J, Sapparapu G, Akahata W, Doranz BJ, Crowe JE, Diamond MS, Rossmann MG. Cryo-EM structures elucidate neutralizing mechanisms of anti-chikungunya human monoclonal antibodies with therapeutic activity. Proc Natl Acad Sci U S A 2015; 112:13898-903. [PMID: 26504196 PMCID: PMC4653152 DOI: 10.1073/pnas.1515558112] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes severe acute and chronic disease in humans. Although highly inhibitory murine and human monoclonal antibodies (mAbs) have been generated, the structural basis of their neutralizing activity remains poorly characterized. Here, we determined the cryo-EM structures of chikungunya virus-like particles complexed with antibody fragments (Fab) of two highly protective human mAbs, 4J21 and 5M16, that block virus fusion with host membranes. Both mAbs bind primarily to sites within the A and B domains, as well as to the B domain's β-ribbon connector of the viral glycoprotein E2. The footprints of these antibodies on the viral surface were consistent with results from loss-of-binding studies using an alanine scanning mutagenesis-based epitope mapping approach. The Fab fragments stabilized the position of the B domain relative to the virus, particularly for the complex with 5M16. This finding is consistent with a mechanism of neutralization in which anti-CHIKV mAbs that bridge the A and B domains impede movement of the B domain away from the underlying fusion loop on the E1 glycoprotein and therefore block the requisite pH-dependent fusion of viral and host membranes.
Collapse
Affiliation(s)
- Feng Long
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | | | - Stephen K Austin
- Departments of Medicine, Molecular Microbiology, Pathology, and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Zhenguo Chen
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Thomas Klose
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Andrei Fokine
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Yue Liu
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Jason Porta
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Gopal Sapparapu
- Department of Pediatrics, Vanderbilt University, Nashville, TN 37232
| | | | | | - James E Crowe
- Department of Pediatrics, Vanderbilt University, Nashville, TN 37232; Departments of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology, and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael G Rossmann
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907;
| |
Collapse
|
71
|
Abdelnabi R, Neyts J, Delang L. Towards antivirals against chikungunya virus. Antiviral Res 2015; 121:59-68. [PMID: 26119058 PMCID: PMC7113767 DOI: 10.1016/j.antiviral.2015.06.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/18/2015] [Accepted: 06/24/2015] [Indexed: 12/25/2022]
Abstract
Chikungunya virus (CHIKV) has re-emerged in recent decades, causing major outbreaks of chikungunya fever in many parts of Africa and Asia, and since the end of 2013 also in Central and South America. Infections are usually associated with a low mortality rate, but can proceed into a painful chronic stage, during which patients may suffer from polyarthralgia and joint stiffness for weeks and even several years. There are no vaccines or antiviral drugs available for the prevention or treatment of CHIKV infections. Current therapy therefore consists solely of the administration of analgesics, antipyretics and anti-inflammatory agents to relieve symptoms. We here review molecules that have been reported to inhibit CHIKV replication, either as direct-acting antivirals, host-targeting drugs or those that act via a yet unknown mechanism. This article forms part of a symposium in Antiviral Research on "Chikungunya discovers the New World."
Collapse
Affiliation(s)
- Rana Abdelnabi
- Rega Institute for Medical Research, University of Leuven, Belgium
| | - Johan Neyts
- Rega Institute for Medical Research, University of Leuven, Belgium.
| | - Leen Delang
- Rega Institute for Medical Research, University of Leuven, Belgium
| |
Collapse
|
72
|
Lam S, Nyo M, Phuektes P, Yew CW, Tan YJ, Chu JJH. A potent neutralizing IgM mAb targeting the N218 epitope on E2 protein protects against Chikungunya virus pathogenesis. MAbs 2015; 7:1178-94. [PMID: 26305993 DOI: 10.1080/19420862.2015.1083664] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Chikungunya virus (CHIKV) is a medically important human viral pathogen that causes Chikungunya fever accompanied with debilitating and persistent joint pain. Host-elicited or passively-transferred monoclonal antibodies (mAb) are essential mediators of CHIKV clearance. Therefore, this study aimed to generate and characterize a panel of mAbs for their neutralization efficacy against CHIKV infection in a cell-based and murine model. To evaluate their antigenicity and neutralization profile, indirect enzyme-linked immunosorbent assay (ELISA), an immunofluorescence assay (IFA) and a plaque reduction neutralization test were performed on mAbs of IgM isotype. CHIKV escape mutants against mAb 3E7b neutralization were generated, and reverse genetics techniques were then used to create an infectious CHIKV clone with a single mutation. 3E7b was also administered to neonate mice prior or after CHIKV infection. The survival rate, CHIKV burden in tissues and histopathology of the limb muscles were evaluated. Both IgM 3E7b and 8A2c bind strongly to native CHIKV surface and potently neutralize CHIKV replication. Further analyses of 3E7b binding and neutralization of CHIKV single-mutant clones revealed that N218 of CHIKV E2 protein is a potent neutralizing epitope. In a pre-binding neutralization assay, 3E7b blocks CHIKV attachment to permissive cells, possibly by binding to the surface-accessible E2-N218 residue. Prophylactic administration of 3E7b to neonate mice markedly reduced viremia and protected against CHIKV pathogenesis in various mice tissues. Given therapeutically at 4 h post-infection, 3E7b conferred 100% survival rate and similarly reduced CHIKV load in most mice tissues except the limb muscles. Collectively, these findings highlight the usefulness of 3E7b for future prophylactic or epitope-based vaccine design.
Collapse
Affiliation(s)
- Shirley Lam
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Min Nyo
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Patchara Phuektes
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Chow Wenn Yew
- b Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research) ; Singapore
| | - Yee Joo Tan
- b Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research) ; Singapore.,c Hepatitis Viruses and Newly Emerging Viruses; Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Justin Jang Hann Chu
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore.,b Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research) ; Singapore
| |
Collapse
|
73
|
Rudd PA, Raphael AP, Yamada M, Nufer KL, Gardner J, Le TTT, Prow NA, Dang N, Schroder WA, Prow TW, Suhrbier A. Effective cutaneous vaccination using an inactivated chikungunya virus vaccine delivered by Foroderm. Vaccine 2015; 33:5172-80. [PMID: 26296498 DOI: 10.1016/j.vaccine.2015.07.099] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 04/14/2015] [Accepted: 07/30/2015] [Indexed: 10/23/2022]
Abstract
Foroderm is a new cutaneous delivery technology that uses high-aspect ratio, cylindrical silica microparticles, that are massaged into the skin using a 3D-printed microtextured applicator, in order to deliver payloads across the epidermis. Herein we show that this technology is effective for delivery of a non-adjuvanted, inactivated, whole-virus chikungunya virus vaccine in mice, with minimal post-vaccination skin reactions. A single topical Foroderm-based vaccination induced T cell, Th1 cytokine and antibody responses, which provided complete protection against viraemia and disease after challenge with chikungunya virus. Foroderm vaccination was shown to deliver fluorescent, virus-sized beads across the epidermis, with beads subsequently detected in draining lymph nodes. Foroderm vaccination also stimulated the egress of MHC II(+) antigen presenting cells from the skin. Foroderm thus has potential as a simple, cheap, effective, generic, needle-free technology for topical delivery of vaccines.
Collapse
Affiliation(s)
- Penny A Rudd
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, QLD, Australia
| | - Anthony P Raphael
- Dermatology Research Centre, University of Queensland, School of Medicine, Translational Research Institute, Brisbane 4102, QLD, Australia
| | - Miko Yamada
- Dermatology Research Centre, University of Queensland, School of Medicine, Translational Research Institute, Brisbane 4102, QLD, Australia
| | - Kaitlin L Nufer
- Dermatology Research Centre, University of Queensland, School of Medicine, Translational Research Institute, Brisbane 4102, QLD, Australia
| | - Joy Gardner
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, QLD, Australia
| | - Thuy T T Le
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, QLD, Australia
| | - Natalie A Prow
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, QLD, Australia; Australian Infectious Disease Research Centre, School of Chemistry & Molecular Biosciences, University of Queensland, Brisbane 4072, QLD, Australia
| | - Nhung Dang
- Dermatology Research Centre, University of Queensland, School of Medicine, Translational Research Institute, Brisbane 4102, QLD, Australia
| | - Wayne A Schroder
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, QLD, Australia
| | - Tarl W Prow
- Dermatology Research Centre, University of Queensland, School of Medicine, Translational Research Institute, Brisbane 4102, QLD, Australia.
| | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, QLD, Australia; Australian Infectious Disease Research Centre, School of Chemistry & Molecular Biosciences, University of Queensland, Brisbane 4072, QLD, Australia
| |
Collapse
|
74
|
Lum FM, Ng LF. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res 2015; 120:165-74. [DOI: 10.1016/j.antiviral.2015.06.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 05/27/2015] [Accepted: 06/16/2015] [Indexed: 12/15/2022]
|
75
|
Smith SA, Silva LA, Fox JM, Flyak AI, Kose N, Sapparapu G, Khomandiak S, Khomadiak S, Ashbrook AW, Kahle KM, Fong RH, Swayne S, Doranz BJ, McGee CE, Heise MT, Pal P, Brien JD, Austin SK, Diamond MS, Dermody TS, Crowe JE. Isolation and Characterization of Broad and Ultrapotent Human Monoclonal Antibodies with Therapeutic Activity against Chikungunya Virus. Cell Host Microbe 2015; 18:86-95. [PMID: 26159721 PMCID: PMC4501771 DOI: 10.1016/j.chom.2015.06.009] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/27/2015] [Accepted: 06/22/2015] [Indexed: 11/25/2022]
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted RNA virus that causes acute febrile infection associated with polyarthralgia in humans. Mechanisms of protective immunity against CHIKV are poorly understood, and no effective therapeutics or vaccines are available. We isolated and characterized human monoclonal antibodies (mAbs) that neutralize CHIKV infectivity. Among the 30 mAbs isolated, 13 had broad and ultrapotent neutralizing activity (IC50 < 10 ng/ml), and all of these mapped to domain A of the E2 envelope protein. Potent inhibitory mAbs blocked post-attachment steps required for CHIKV membrane fusion, and several were protective in a lethal challenge model in immunocompromised mice, even when administered at late time points after infection. These highly protective mAbs could be considered for prevention or treatment of CHIKV infection, and their epitope location in domain A of E2 could be targeted for rational structure-based vaccine development.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/isolation & purification
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/immunology
- Antibodies, Viral/isolation & purification
- Antibodies, Viral/therapeutic use
- Chemoprevention/methods
- Chikungunya Fever/therapy
- Chikungunya virus/immunology
- Chikungunya virus/physiology
- Disease Models, Animal
- Humans
- Immunization, Passive/methods
- Inhibitory Concentration 50
- Mice
- Protein Binding
- Survival Analysis
- Treatment Outcome
- Viral Envelope Proteins/immunology
- Virus Internalization/drug effects
Collapse
Affiliation(s)
- Scott A Smith
- Department of Medicine, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Laurie A Silva
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Julie M Fox
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew I Flyak
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Gopal Sapparapu
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Solomiia Khomadiak
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Alison W Ashbrook
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | | | - Charles E McGee
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Mark T Heise
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Pankaj Pal
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James D Brien
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - S Kyle Austin
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Terence S Dermody
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - James E Crowe
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
76
|
Abstract
Chikungunya virus (CHIKV) is a rapidly emerging mosquito-borne alphavirus causing millions of infections in the tropical and subtropical regions of the world. CHIKV infection often leads to an acute self-limited febrile illness with debilitating myalgia and arthralgia. A potential long-term complication of CHIKV infection is severe joint pain, which can last for months to years. There are no vaccines or specific therapeutics available to prevent or treat infection. This review describes the critical steps in CHIKV cell entry. We summarize the latest studies on the virus-cell tropism, virus-receptor binding, internalization, membrane fusion and review the molecules and compounds that have been described to interfere with virus cell entry. The aim of the review is to give the reader a state-of-the-art overview on CHIKV cell entry and to provide an outlook on potential new avenues in CHIKV research.
Collapse
|
77
|
Couderc T, Lecuit M. Chikungunya virus pathogenesis: From bedside to bench. Antiviral Res 2015; 121:120-31. [PMID: 26159730 DOI: 10.1016/j.antiviral.2015.07.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/04/2015] [Indexed: 11/28/2022]
Abstract
Chikungunya virus (CHIKV) is an arbovirus transmitted to humans by mosquito bite. A decade ago, the virus caused a major outbreak in the islands of the Indian Ocean, then reached India and Southeast Asia. More recently, CHIKV has emerged in the Americas, first reaching the Caribbean and now extending to Central, South and North America. It is therefore considered a major public health and economic threat. CHIKV causes febrile illness typically associated with debilitating joint pains. In rare cases, it may also cause central nervous system disease, notably in neonates. Joint symptoms may persist for months to years, and lead to arthritis. This review focuses on the spectrum of signs and symptoms associated with CHIKV infection in humans. It also illustrates how the analysis of clinical and biological data from human cohorts and the development of animal and cellular models of infection has helped to identify the tissue and cell tropisms of the virus and to decipher host responses in benign, severe or persistent disease. This article forms part of a symposium in Antiviral Research on "Chikungunya discovers the New World".
Collapse
Affiliation(s)
- Thérèse Couderc
- Institut Pasteur, Biology of Infection Unit, Paris, France; Inserm U1117, Paris, France.
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France; Inserm U1117, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Division of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades University Hospital, Institut Imagine, Paris, France; Global Virus Network.
| |
Collapse
|
78
|
Poo YS, Rudd PA, Gardner J, Wilson JAC, Larcher T, Colle MA, Le TT, Nakaya HI, Warrilow D, Allcock R, Bielefeldt-Ohmann H, Schroder WA, Khromykh AA, Lopez JA, Suhrbier A. Multiple immune factors are involved in controlling acute and chronic chikungunya virus infection. PLoS Negl Trop Dis 2014; 8:e3354. [PMID: 25474568 PMCID: PMC4256279 DOI: 10.1371/journal.pntd.0003354] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 10/15/2014] [Indexed: 12/18/2022] Open
Abstract
The recent epidemic of the arthritogenic alphavirus, chikungunya virus (CHIKV) has prompted a quest to understand the correlates of protection against virus and disease in order to inform development of new interventions. Herein we highlight the propensity of CHIKV infections to persist long term, both as persistent, steady-state, viraemias in multiple B cell deficient mouse strains, and as persistent RNA (including negative-strand RNA) in wild-type mice. The knockout mouse studies provided evidence for a role for T cells (but not NK cells) in viraemia suppression, and confirmed the role of T cells in arthritis promotion, with vaccine-induced T cells also shown to be arthritogenic in the absence of antibody responses. However, MHC class II-restricted T cells were not required for production of anti-viral IgG2c responses post CHIKV infection. The anti-viral cytokines, TNF and IFNγ, were persistently elevated in persistently infected B and T cell deficient mice, with adoptive transfer of anti-CHIKV antibodies unable to clear permanently the viraemia from these, or B cell deficient, mice. The NOD background increased viraemia and promoted arthritis, with B, T and NK deficient NOD mice showing high-levels of persistent viraemia and ultimately succumbing to encephalitic disease. In wild-type mice persistent CHIKV RNA and negative strand RNA (detected for up to 100 days post infection) was associated with persistence of cellular infiltrates, CHIKV antigen and stimulation of IFNα/β and T cell responses. These studies highlight that, secondary to antibodies, several factors are involved in virus control, and suggest that chronic arthritic disease is a consequence of persistent, replicating and transcriptionally active CHIKV RNA. The largest epidemic ever recorded for chikungunya virus (CHIKV) started in 2004 in Africa, then spread across Asia and recently caused tens of thousands of cases in Papua New Guinea and the Caribbean. This mosquito-borne alphavirus primarily causes an often debilitating, acute and chronic polyarthritis/polyarthalgia. Despite robust anti-viral immune responses CHIKV is able to persist, with such persistence poorly understood and the likely cause of chronic disease. Herein we highlight the propensity of CHIKV to persist long term, both as a persistent viraemia in different B cell deficient mouse strains, but also as persistent viral RNA in wild-type mice. These studies suggest that, aside from antibodies, other immune factors, such as CD4 T cells and TNF, are active in viraemia control. The work also supports the notion that CHIKV disease, with the exception of encephalitis, is largely an immunopathology. Persistent CHIKV RNA in wild-type mice continues to stimulate type I interferon and T cell responses, with this model of chronic disease recapitulating many of the features seen in chronic CHIKV patients.
Collapse
Affiliation(s)
- Yee Suan Poo
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Penny A. Rudd
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Joy Gardner
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
| | - Jane A. C. Wilson
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Thibaut Larcher
- Institut National de Recherche Agronomique, Unité Mixte de Recherche 703, Oniris, Nantes, France
| | - Marie-Anne Colle
- Institut National de Recherche Agronomique, Unité Mixte de Recherche 703, Oniris, Nantes, France
| | - Thuy T. Le
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
| | - Helder I. Nakaya
- School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - David Warrilow
- Public Health Virology Laboratory, Department of Health, Queensland Government, Brisbane, Queensland, Australia
| | - Richard Allcock
- Lotterywest State Biomedical Facility Genomics, Royal Perth Hospital, Perth, Western Australia, Australia
| | | | - Wayne A. Schroder
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
| | - Alexander A. Khromykh
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - José A. Lopez
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Natural Sciences, Griffith University, Nathan, Australia
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
- School of Natural Sciences, Griffith University, Nathan, Australia
- * E-mail:
| |
Collapse
|
79
|
Exposure of epitope residues on the outer face of the chikungunya virus envelope trimer determines antibody neutralizing efficacy. J Virol 2014; 88:14364-79. [PMID: 25275138 DOI: 10.1128/jvi.01943-14] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Chikungunya virus (CHIKV) is a reemerging alphavirus that causes a debilitating arthritic disease and infects millions of people and for which no specific treatment is available. Like many alphaviruses, the structural targets on CHIKV that elicit a protective humoral immune response in humans are poorly defined. Here we used phage display against virus-like particles (VLPs) to isolate seven human monoclonal antibodies (MAbs) against the CHIKV envelope glycoproteins E2 and E1. One MAb, IM-CKV063, was highly neutralizing (50% inhibitory concentration, 7.4 ng/ml), demonstrated high-affinity binding (320 pM), and was capable of therapeutic and prophylactic protection in multiple animal models up to 24 h postexposure. Epitope mapping using a comprehensive shotgun mutagenesis library of 910 mutants with E2/E1 alanine mutations demonstrated that IM-CKV063 binds to an intersubunit conformational epitope on domain A, a functionally important region of E2. MAbs against the highly conserved fusion loop have not previously been reported but were also isolated in our studies. Fusion loop MAbs were broadly cross-reactive against diverse alphaviruses but were nonneutralizing. Fusion loop MAb reactivity was affected by temperature and reactivity conditions, suggesting that the fusion loop is hidden in infectious virions. Visualization of the binding sites of 15 different MAbs on the structure of E2/E1 revealed that all epitopes are located at the membrane-distal region of the E2/E1 spike. Interestingly, epitopes on the exposed topmost and outer surfaces of the E2/E1 trimer structure were neutralizing, whereas epitopes facing the interior of the trimer were not, providing a rationale for vaccine design and therapeutic MAb development using the intact CHIKV E2/E1 trimer. IMPORTANCE CHIKV is the most important alphavirus affecting humans, resulting in a chronic arthritic condition that can persist for months or years. In recent years, millions of people have been infected globally, and the spread of CHIKV to the Americas is now beginning, with over 100,000 cases occurring in the Caribbean within 6 months of its arrival. Our study reports on seven human MAbs against the CHIKV envelope, including a highly protective MAb and rarely isolated fusion loop MAbs. Epitope mapping of these MAbs demonstrates how some E2/E1 epitopes are exposed or hidden from the human immune system and suggests a structural mechanism by which these MAbs protect (or fail to protect) against CHIKV infection. Our results suggest that the membrane-distal end of CHIKV E2/E1 is the primary target for the humoral immune response to CHIKV, and antibodies targeting the exposed topmost and outer surfaces of the E2/E1 trimer determine the neutralizing efficacy of this response.
Collapse
|
80
|
Wisskirchen K, Lucifora J, Michler T, Protzer U. New pharmacological strategies to fight enveloped viruses. Trends Pharmacol Sci 2014; 35:470-8. [PMID: 25108320 PMCID: PMC7112871 DOI: 10.1016/j.tips.2014.06.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/13/2014] [Accepted: 06/17/2014] [Indexed: 02/07/2023]
Abstract
Many emerging or known, chronic viral diseases are caused by enveloped viruses. The review discusses research driven development of antivirals that became recently available or are in clinical evaluation. The main focus is on antiviral strategies with a broader therapeutic range, and on novel immune based therapeutics. Broad-spectrum antivirals will help to react faster to newly emerging viral diseases. Targeting immune cells against infected cells can restore immune responses in chronic infections.
Enveloped viruses pose an important health threat because most of the persistent and many emerging viruses are enveloped. In particular, newly emerging viruses create a need to develop broad-spectrum antivirals, which usually are obtained by targeting host cell factors. Persistent viruses have developed efficient strategies to escape host immune control, and treatment options are limited. Targeting host cell factors essential for virus persistence, or immune-based therapies provide alternative approaches. In this review, we therefore focus on recent developments to generate antivirals targeting host cell factors or immune-based therapeutic approaches to fight infections with enveloped viruses.
Collapse
Affiliation(s)
- Karin Wisskirchen
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Trogerstrasse 30, 81675 Munich, Germany
| | - Julie Lucifora
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Trogerstrasse 30, 81675 Munich, Germany; German Center for Infection Research (DZIF), Munich site, Munich, Germany
| | - Thomas Michler
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Trogerstrasse 30, 81675 Munich, Germany; German Center for Infection Research (DZIF), Munich site, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Trogerstrasse 30, 81675 Munich, Germany; German Center for Infection Research (DZIF), Munich site, Munich, Germany.
| |
Collapse
|
81
|
Chikungunya viruses that escape monoclonal antibody therapy are clinically attenuated, stable, and not purified in mosquitoes. J Virol 2014; 88:8213-26. [PMID: 24829346 DOI: 10.1128/jvi.01032-14] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Chikungunya virus (CHIKV) is a reemerging mosquito-transmitted alphavirus that causes epidemics of debilitating polyarthritis in humans. A prior study identified two anti-CHIKV monoclonal antibodies ([MAbs] CHK-152 and CHK-166) against the E2 and E1 structural proteins, which had therapeutic efficacy in immunocompetent and immunocompromised mice. Combination MAb therapy was required as administration of a single MAb resulted in the rapid selection of neutralization escape variants and treatment failure in mice. Here, we initially evaluated the efficacy of combination MAb therapy in a nonhuman primate model of CHIKV infection. Treatment of rhesus macaques with CHK-152 and CHK-166 reduced viral spread and infection in distant tissue sites and also neutralized reservoirs of infectious virus. Escape viruses were not detected in the residual viral RNA present in tissues and organs of rhesus macaques. To evaluate the possible significance of MAb resistance, we engineered neutralization escape variant viruses (E1-K61T, E2-D59N, and the double mutant E1-K61T E2-D59N) that conferred resistance to CHK-152 and CHK-166 and tested them for fitness in mosquito cells, mammalian cells, mice, and Aedes albopictus mosquitoes. In both cell culture and mosquitoes, the mutant viruses grew equivalently and did not revert to wild-type (WT) sequence. All escape variants showed evidence of mild clinical attenuation, with decreased musculoskeletal disease at early times after infection in WT mice and a prolonged survival time in immunocompromised Ifnar1(-/-) mice. Unexpectedly, this was not associated with decreased infectivity, and consensus sequencing from tissues revealed no evidence of reversion or compensatory mutations. Competition studies with CHIKV WT also revealed no fitness compromise of the double mutant (E1-K61T E2-D59N) neutralization escape variant in WT mice. Collectively, our study suggests that neutralization escape viruses selected during combination MAb therapy with CHK-152 plus CHK-166 retain fitness, cause less severe clinical disease, and likely would not be purified during the enzootic cycle. IMPORTANCE Chikungunya virus (CHIKV) causes explosive epidemics of acute and chronic arthritis in humans in Africa, the Indian subcontinent, and Southeast Asia and recently has spread to the New World. As there are no approved vaccines or therapies for human use, the possibility of CHIKV-induced debilitating disease is high in many parts of the world. To this end, our laboratory recently generated a combination monoclonal antibody therapy that aborted lethal and arthritogenic disease in wild-type and immunocompromised mice when administered as a single dose several days after infection. In this study, we show the efficacy of the antibody combination in nonhuman primates and also evaluate the significance of possible neutralization escape mutations in mosquito and mammalian cells, mice, and Aedes albopictus vector mosquitoes. Our experiments show that escape viruses from combination antibody therapy cause less severe CHIKV clinical disease, retain fitness, and likely would not be purified by mosquito vectors.
Collapse
|
82
|
Kam YW, Lee WWL, Simarmata D, Le Grand R, Tolou H, Merits A, Roques P, Ng LFP. Unique epitopes recognized by antibodies induced in Chikungunya virus-infected non-human primates: implications for the study of immunopathology and vaccine development. PLoS One 2014; 9:e95647. [PMID: 24755730 PMCID: PMC3995782 DOI: 10.1371/journal.pone.0095647] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 03/28/2014] [Indexed: 11/17/2022] Open
Abstract
Chikungunya virus (CHIKV) is an Alphavirus that causes chronic and incapacitating arthralgia in humans. Although patient cohort studies have shown the production of CHIKV specific antibodies, the fine specificity of the antibody response against CHIKV is not completely defined. The macaque model of CHIKV infection was established due to limitations of clinical specimens. More importantly, its close relation to humans will allow the study of chronic infection and further identify important CHIKV targets. In this study, serum samples from CHIKV-infected macaques collected at different time-points post infection were used to characterize the antibody production pattern and kinetics. Results revealed that anti-CHIKV antibodies were neutralizing and the E2 glycoprotein, Capsid, nsP1, nsP3 and nsP4 proteins were targets of the anti-CHIKV antibody response in macaques. Furthermore, linear B-cell epitopes recognized by these anti-CHIKV antibodies were identified, and mapped to their structural localization. This characterizes the specificity of anti-CHIKV antibody response in macaques and further demonstrates the importance of the different regions in CHIKV-encoded proteins in the adaptive immune response. Information from this study provides critical knowledge that will aid in the understanding of CHIKV infection and immunity, vaccine design, and pre-clinical efficacy studies.
Collapse
Affiliation(s)
- Yiu-Wing Kam
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Wendy W L Lee
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Diane Simarmata
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Roger Le Grand
- CEA, Division of ImmunoVirology (SIV), Institute of Emerging Diseases and Innovative Therapies (IMETI), Fontenay-aux-Roses, France; Université Paris-Sud 11, UMR E1, Orsay, France
| | - Hugues Tolou
- Groupe d'Etude en Préventologie (GEP), Villenave d'Ornon, France
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Pierre Roques
- CEA, Division of ImmunoVirology (SIV), Institute of Emerging Diseases and Innovative Therapies (IMETI), Fontenay-aux-Roses, France; Université Paris-Sud 11, UMR E1, Orsay, France
| | - Lisa F P Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
83
|
A novel poxvirus-based vaccine, MVA-CHIKV, is highly immunogenic and protects mice against chikungunya infection. J Virol 2014; 88:3527-47. [PMID: 24403588 DOI: 10.1128/jvi.03418-13] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED There is a need to develop a single and highly effective vaccine against the emerging chikungunya virus (CHIKV), which causes a severe disease in humans. Here, we have generated and characterized the immunogenicity profile and the efficacy of a novel CHIKV vaccine candidate based on the highly attenuated poxvirus vector modified vaccinia virus Ankara (MVA) expressing the CHIKV C, E3, E2, 6K, and E1 structural genes (termed MVA-CHIKV). MVA-CHIKV was stable in cell culture, expressed the CHIKV structural proteins, and triggered the cytoplasmic accumulation of Golgi apparatus-derived membranes in infected human cells. Furthermore, MVA-CHIKV elicited robust innate immune responses in human macrophages and monocyte-derived dendritic cells, with production of beta interferon (IFN-β), proinflammatory cytokines, and chemokines. After immunization of C57BL/6 mice with a homologous protocol (MVA-CHIKV/MVA-CHIKV), strong, broad, polyfunctional, and durable CHIKV-specific CD8(+) T cell responses were elicited. The CHIKV-specific CD8(+) T cells were preferentially directed against E1 and E2 proteins and, to a lesser extent, against C protein. CHIKV-specific CD8(+) memory T cells of a mainly effector memory phenotype were also induced. The humoral arm of the immune system was significantly induced, as MVA-CHIKV elicited high titers of neutralizing antibodies against CHIKV. Remarkably, a single dose of MVA-CHIKV protected all mice after a high-dose challenge with CHIKV. In summary, MVA-CHIKV is an effective vaccine against chikungunya virus infection that induced strong, broad, highly polyfunctional, and long-lasting CHIKV-specific CD8(+) T cell responses, together with neutralizing antibodies against CHIKV. These results support the consideration of MVA-CHIKV as a potential vaccine candidate against CHIKV. IMPORTANCE We have developed a novel vaccine candidate against chikungunya virus (CHIKV) based on the highly attenuated poxvirus vector modified vaccinia virus Ankara (MVA) expressing the CHIKV C, E3, E2, 6K, and E1 structural genes (termed MVA-CHIKV). Our findings revealed that MVA-CHIKV is a highly effective vaccine against chikungunya virus, with a single dose of the vaccine protecting all mice after a high-dose challenge with CHIKV. Furthermore, MVA-CHIKV is highly immunogenic, inducing strong innate responses: high, broad, polyfunctional, and long-lasting CHIKV-specific CD8(+) T cell responses, together with neutralizing antibodies against CHIKV. This work provides a potential vaccine candidate against CHIKV.
Collapse
|