1
|
Riccardo V, Pablo GC. Neutralization Determinants on Poxviruses. Viruses 2023; 15:2396. [PMID: 38140637 PMCID: PMC10747254 DOI: 10.3390/v15122396] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Smallpox was a highly contagious disease caused by the variola virus. The disease affected millions of people over thousands of years and variola virus ranked as one of the deadliest viruses in human history. The complete eradication of smallpox in 1980, a major triumph in medicine, was achieved through a global vaccination campaign using a less virulent poxvirus, vaccinia virus. Despite this success, the herd immunity established by this campaign has significantly waned, and concerns are rising about the potential reintroduction of variola virus as a biological weapon or the emergence of zoonotic poxviruses. These fears were further fueled in 2022 by a global outbreak of monkeypox virus (mpox), which spread to over 100 countries, thereby boosting interest in developing new vaccines using molecular approaches. However, poxviruses are complex and creating modern vaccines against them is challenging. This review focuses on the structural biology of the six major neutralization determinants on poxviruses (D8, H3, A27, L1, B5, and A33), the localization of epitopes targeted by neutralizing antibodies, and their application in the development of subunit vaccines.
Collapse
Affiliation(s)
| | - Guardado-Calvo Pablo
- Structural Biology of Infectious Diseases Unit, Institut Pasteur, Université Paris Cité, F-75015 Paris, France;
| |
Collapse
|
2
|
Leão TL, Lourenço KL, de Oliveira Queiroz C, Serufo ÂV, da Silva AM, Barbosa-Stancioli EF, da Fonseca FG. Vaccinia virus induces endoplasmic reticulum stress and activates unfolded protein responses through the ATF6α transcription factor. Virol J 2023; 20:145. [PMID: 37434252 DOI: 10.1186/s12985-023-02122-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/08/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Cell responses to different stress inducers are efficient mechanisms that prevent and fight the accumulation of harmful macromolecules in the cells and also reinforce the defenses of the host against pathogens. Vaccinia virus (VACV) is an enveloped, DNA virus, belonging to the Poxviridae family. Members of this family have evolved numerous strategies to manipulate host responses to stress controlling cell survival and enhancing their replicative success. In this study, we investigated the activation of the response signaling to malformed proteins (UPR) by the VACV virulent strain-Western Reserve (WR)-or the non-virulent strain-Modified Vaccinia Ankara (MVA). METHODS Through RT-PCR RFLP and qPCR assays, we detected negative regulation of XBP1 mRNA processing in VACV-infected cells. On the other hand, through assays of reporter genes for the ATF6 component, we observed its translocation to the nucleus of infected cells and a robust increase in its transcriptional activity, which seems to be important for virus replication. WR strain single-cycle viral multiplication curves in ATF6α-knockout MEFs showed reduced viral yield. RESULTS We observed that VACV WR and MVA strains modulate the UPR pathway, triggering the expression of endoplasmic reticulum chaperones through ATF6α signaling while preventing IRE1α-XBP1 activation. CONCLUSIONS The ATF6α sensor is robustly activated during infection while the IRE1α-XBP1 branch is down-regulated.
Collapse
Affiliation(s)
- Thiago Lima Leão
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Karine Lima Lourenço
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Cid de Oliveira Queiroz
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Ângela Vieira Serufo
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Aristóbolo Mendes da Silva
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Edel F Barbosa-Stancioli
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Flávio Guimarães da Fonseca
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| |
Collapse
|
3
|
Abstract
INTRODUCTION In May 2022, the World Health Organisation declared a multi-country monkeypox outbreak in non-endemic countries following cases reported from 12 member states that were not endemic for monkeypox virus. SOURCES OF DATA Pubmed search. AREAS OF AGREEMENT The virology, epidemiology, transmission, incubation and aspects of infection control are described. Clinical features of previous and current outbreaks are described, with growing observations that the current outbreak presents with clinical features distinct from previous outbreaks. AREAS OF CONTROVERSY There are variations in clinical presentations seen in the current outbreak that have not been seen in prior outbreaks. More research is needed to investigate the reasons for these differences. GROWING POINTS The higher numbers of HIV-positive patients in the current outbreak has allowed better description of the disease in patients co-infected with HIV and monkeypox. The absence of more severe symptoms in HIV-positive patients in the current outbreak could possibly be due to the fact that most of these patients had well-controlled HIV, although further characterization of this cohort of patients would be useful. AREAS FOR DEVELOPING RESEARCH Current treatment and vaccination options have been extrapolated from studies of other Orthopox viruses. There remains a need for more data on the safety and efficacy of these options in the context of monkeypox infections.
Collapse
Affiliation(s)
- Eleanor Y Lim
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - James Whitehorn
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK.,Clinical Microbiology and Public Health Laboratory, Public Health England, Cambridge CB21 5XA, UK
| | - Lucy Rivett
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK.,Clinical Microbiology and Public Health Laboratory, Public Health England, Cambridge CB21 5XA, UK
| |
Collapse
|
4
|
Shafaati M, Zandi M. Human monkeypox (hMPXV) re-emergence: Host immunity status and current vaccines landscape. J Med Virol 2023; 95:e28251. [PMID: 36271768 DOI: 10.1002/jmv.28251] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/28/2022] [Accepted: 10/19/2022] [Indexed: 01/11/2023]
Abstract
Monkeypox virus is a member of the Orthopoxvirus genus and the Poxviridae family. Orthopoxviruses are among the most intricate animal viruses. The pathogenicity of human monkeypox infection has been emphasized in response to its recent emergence in non-endemic countries and the threat of bioterrorism. It is always necessary to take appropriate precautions in exposure to emerging or re-emerging infections. Here, we focus on the current state of the human monkeypox infection outbreak, research & development of immune responses, and clinical interventions to prevent and treat the human monkeypox virus and other human poxviruses.
Collapse
Affiliation(s)
- Maryam Shafaati
- Department of Microbiology, Faculty of Science, Jahrom Branch, Islamic Azad University, Jahrom, Iran
- Occupational Sleep Research, Baharloo Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Milad Zandi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Fay P, Limon G, Ulziibat G, Khanui B, Myagmarsuren O, Tore G, Khishgee B, Flannery J, Sandag B, Damdinjav B, Beard PM. A field study evaluating the humoral immune response in Mongolian sheep vaccinated against sheeppox virus. Transbound Emerg Dis 2022; 69:1837-1846. [PMID: 34033248 DOI: 10.1111/tbed.14163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/04/2021] [Accepted: 05/20/2021] [Indexed: 12/20/2022]
Abstract
Sheeppox is a transboundary disease of small ruminants caused by infection with the capripoxvirus sheeppox virus. Sheeppox is found in Africa, the Middle East and Asia and is characterized by fever, multifocal cutaneous raised lesions and death. Vaccination with live attenuated capripoxvirus (CPPV) strains is an effective and widely used strategy to contol sheeppox outbreaks; however, there are few reports of post-vaccination field surveillance studies. This study used a commercially available enzyme-linked immunosorbent assay (ELISA) to examine quantitative and temporal features of the humoral response of sheep vaccinated with a live-attenuated CPPV strain in Mongolia. Four hundred samples were tested using the ELISA commercial kit, and a subset of 45 samples were also tested with a virus neutralization test (VNT). There was substantial agreement between the VNT and ELISA tests. Antibodies to CPPV were detected between 40 and 262 days post-vaccination. There was no significant difference between serological status (positive/negative) and sex or age; however, an inverse correlation was found between the length of time since vaccination and serological status. Animals between 90 and 180 days post-vaccination were more likely to be positive than animals greater than 180 days post-vaccination. Our results show that a commercial CPPV ELISA kit is a robust and reliable assay for post-CPPV vaccination surveillance in resource-restricted settings and provide temporal parameters to be considered when planning sheeppox post-vaccination monitoring programmes.
Collapse
Affiliation(s)
- Petra Fay
- The Pirbright Institute, Pirbright, UK
| | | | - Gerelmaa Ulziibat
- State Central Veterinary Laboratory, Zaisan, Khan-Uul District, Ulaanbaatar, Mongolia
| | - Buyantogtokh Khanui
- State Central Veterinary Laboratory, Zaisan, Khan-Uul District, Ulaanbaatar, Mongolia
| | | | | | - Bodisaikhan Khishgee
- State Central Veterinary Laboratory, Zaisan, Khan-Uul District, Ulaanbaatar, Mongolia
| | | | - Batkhuyag Sandag
- The Mongolian General Authority for Veterinary Services, Ulaanbaatar, Mongolia
| | - Batchuluun Damdinjav
- State Central Veterinary Laboratory, Zaisan, Khan-Uul District, Ulaanbaatar, Mongolia
| | - Philippa M Beard
- The Pirbright Institute, Pirbright, UK.,The Roslin Institute, Easter Bush, University of Edinburgh, UK
| |
Collapse
|
6
|
Kennedy RB, Ovsyannikova IG, Haralambieva IH, Grill DE, Poland GA. Proteomic assessment of humoral immune responses in smallpox vaccine recipients. Vaccine 2022; 40:789-797. [PMID: 34952760 PMCID: PMC8792332 DOI: 10.1016/j.vaccine.2021.12.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 11/09/2021] [Accepted: 12/13/2021] [Indexed: 02/02/2023]
Abstract
The availability of effective smallpox vaccines was a critical element of the successful eradication of smallpox in 1980. Antibody responses play a primary role in protective immunity and neutralizing antibody is an established correlate of protection against smallpox. In this study we used a poxvirus proteome array to assess the antibody response to individual viral proteins in a cohort of 1,037 smallpox vaccine recipients. Several statistically significant differences were observed in the antibody response to immunodominant proteins between men and women, including B5R-a major target of neutralizing antibody in vaccinia immune globulin, and the membrane proteins D8L and A27L, both of which have been used as vaccine antigens providing protection in animal models. We also noted differences across racial/ethnic groups. In this cohort, which consisted of both ACAM2000 and Dryvax recipients, we noted minute differences in the antibody responses to a restricted number of viral proteins, providing additional support for the use of ACAM2000 as a replacement smallpox vaccine. Furthermore, our data indicate that poxvirus proteome microarrays can be valuable for screening and monitoring smallpox vaccine-induced humoral immune responses in large-scale serologic surveillance studies and prove useful in the guidance of developing novel smallpox candidate vaccines.
Collapse
Affiliation(s)
- Richard B. Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN USA,Department of Internal Medicine, Mayo Clinic, Rochester, MN USA,Corresponding author: Richard B. Kennedy, Ph.D., Co-Director, Mayo Vaccine Research Group, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, Phone: (507) 284-0708, Fax: (507) 266-4716,
| | - Inna G. Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN USA,Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Iana H. Haralambieva
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN USA,Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Diane E. Grill
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
| | - Gregory A. Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN USA,Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
7
|
Delacruz WP, Savona MR, Thornton JA, Danaher PJ. Evidence of vaccinia dissemination despite lack of major reaction following smallpox vaccination. Vaccine 2019; 38:1589-1592. [PMID: 31899026 DOI: 10.1016/j.vaccine.2019.12.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 10/25/2022]
Abstract
Following vaccinia vaccination, vesicle formation at the site occurs in 95% of primary vaccinees and is thought to indicate virus replication and vaccine efficacy. Little is known about virus replication and immune response in those who do not develop a vesicle. We used PCR to detect vaccinia in various sites following receipt of the smallpox vaccine in those with and without vesicle formation. Among 80 participants, 74 developed and 6 failed to develop a vesicle. Vaccinia DNA was detected in the blood, in the oropharynx, on the dressing, and on the hands of 5%, 11%, 4%, and 0% of those with vesicle formation and of 33%, 17%, 0%, and 17% of those without vesicle formation, respectively (p > 0.05 for each site). The detection of systemic vaccinia DNA in vaccinees without vesicle formation challenges the current understanding that lack of vesicle formation indicates lack of virus replication, the prerequisite to immune response.
Collapse
Affiliation(s)
- Wilfred P Delacruz
- Clinical Investigation Facility, David Grant United States Air Force Medical Center, 101 Bodin Circle, Travis AFB, CA 94535, USA.
| | - Michael R Savona
- Department of Medicine, David Grant United States Air Force Medical Center, 101 Bodin Circle, Travis AFB, CA 94535, USA.
| | - Jennifer A Thornton
- Clinical Investigation Facility, David Grant United States Air Force Medical Center, 101 Bodin Circle, Travis AFB, CA 94535, USA.
| | - Patrick J Danaher
- Department of Medicine, David Grant United States Air Force Medical Center, 101 Bodin Circle, Travis AFB, CA 94535, USA.
| |
Collapse
|
8
|
Nakatake M, Kurosaki H, Kuwano N, Horita K, Ito M, Kono H, Okamura T, Hasegawa K, Yasutomi Y, Nakamura T. Partial Deletion of Glycoprotein B5R Enhances Vaccinia Virus Neutralization Escape while Preserving Oncolytic Function. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:159-171. [PMID: 31236440 PMCID: PMC6580015 DOI: 10.1016/j.omto.2019.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 05/09/2019] [Indexed: 11/12/2022]
Abstract
Vaccinia virus (VV) has been utilized in oncolytic virotherapy, but it risks a host antiviral immune response. VV has an extracellular enveloped virus (EEV) form consisting of a normal virion covered with a host-derived outer membrane that enables its spread via circulation while evading host immune mechanisms. However, the immune resistance of EEV is only partial, owing to expression of the surface protein B5R, which has four short consensus repeat (SCR) domains that are targeted by host immune factors. To engineer a more effective virus for oncolytic virotherapy, we developed an enhanced immune-evading oncolytic VV by removing the SCRs from the attenuated strain LC16mO. Although deletion of only the SCRs preserved viral replication, progeny production, and oncolytic activity, deletion of whole B5R led to attenuation of the virus. Importantly, SCR-deleted EEV had higher neutralization resistance than did B5R-wild-type EEV against VV-immunized animal serum; moreover, it retained oncolytic function, thereby prolonging the survival of tumor-bearing mice treated with anti-VV antibody. These results demonstrate that partial SCR deletion increases neutralization escape without affecting the oncolytic potency of VV, making it useful for the treatment of tumors under the anti-virus antibody existence.
Collapse
Affiliation(s)
- Motomu Nakatake
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Hajime Kurosaki
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Nozomi Kuwano
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Kosuke Horita
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Mai Ito
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Hiromichi Kono
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Tomotaka Okamura
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki 305-0843, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka-City, Saitama 350-1298, Japan
| | - Yasuhiro Yasutomi
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki 305-0843, Japan
| | - Takafumi Nakamura
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| |
Collapse
|
9
|
Cheminay C, Körner J, Bernig C, Brückel M, Feigl M, Schletz M, Suter M, Chaplin P, Volkmann A. A single vaccination with non-replicating MVA at birth induces both immediate and long-term protective immune responses. Vaccine 2018; 36:2427-2434. [PMID: 29599088 DOI: 10.1016/j.vaccine.2018.03.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/17/2017] [Accepted: 03/16/2018] [Indexed: 11/16/2022]
Abstract
Newborns are considered difficult to protect against infections shortly after birth, due to their ineffective immune system that shows quantitative and qualitative differences compared to adults. However, here we show that a single vaccination of mice at birth with a replication-deficient live vaccine Modified Vaccinia Ankara [MVA] efficiently induces antigen-specific B- and T-cells that fully protect against a lethal Ectromelia virus challenge. Protection was induced within 2 weeks and using genetically modified mice we show that this protection was mainly T-cell dependent. Persisting immunological T-cell memory and neutralizing antibodies were obtained with the single vaccination. Thus, MVA administered as early as at birth induced immediate and long-term protection against an otherwise fatal disease and appears attractive as a new generation smallpox vaccine that is effective also in children. Moreover, it may have the potential to serve as platform for childhood vaccines as indicated by measles specific T- and B-cell responses induced in newborn mice vaccinated with recombinant MVA expressing measles antigens.
Collapse
Affiliation(s)
- Cédric Cheminay
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Jana Körner
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Constanze Bernig
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Michael Brückel
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Markus Feigl
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Martin Schletz
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Mark Suter
- University of Zürich, Dekanat Vetsuisse-Fakultät Immunology, Winterthurerstrasse 204, CH-8057 Zürich, Switzerland
| | - Paul Chaplin
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Ariane Volkmann
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany.
| |
Collapse
|
10
|
Increased attenuation but decreased immunogenicity by deletion of multiple vaccinia virus immunomodulators. Vaccine 2016; 34:4827-34. [PMID: 27544585 PMCID: PMC5022402 DOI: 10.1016/j.vaccine.2016.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/30/2016] [Accepted: 08/01/2016] [Indexed: 12/18/2022]
Abstract
Vaccinia virus-derived vaccine vectors are being engineered to improve immunogenicity. Deleting genes with immunomodulatory function can increase immunogenicity and decrease virulence. Deletion of N1L, C6L or K7R individually improves immunogenicity, but not in combination. A virus lacking all three genes induces poorer CD8+ T cell and neutralising antibody responses.
Vaccinia virus (VACV)-derived vectors are popular candidates for vaccination against diseases such as HIV-1, malaria and tuberculosis. However, their genomes encode a multitude of proteins with immunomodulatory functions, several of which reduce the immunogenicity of these vectors. Hitherto only limited studies have investigated whether the removal of these immunomodulatory genes in combination can increase vaccine efficacy further. To this end we constructed viruses based on VACV strain Western Reserve (WR) lacking up to three intracellular innate immunomodulators (N1, C6 and K7). These genes were selected because the encoded proteins had known functions in innate immunity and the deletion of each gene individually had caused a decrease in virus virulence in the murine intranasal and intradermal models of infection and an increase in immunogenicity. Data presented here demonstrate that deletion of two, or three of these genes in combination attenuated the virus further in an incremental manner. However, when vaccinated mice were challenged with VACV WR the double and triple gene deletion viruses provided weaker protection against challenge. This was accompanied by inferior memory CD8+ T cell responses and lower neutralising antibody titres. This study indicates that, at least for the three genes studied in the context of VACV WR, the single gene deletion viruses are the best vaccine vectors, and that increased attenuation induced by deletion of additional genes decreased immunogenicity. These data highlight the fine balance and complex relationship between viral attenuation and immunogenicity. Given that the proteins encoded by the genes examined in this study are known to affect specific aspects of innate immunity, the set of viruses constructed here are interesting tools to probe the role of the innate immune response in influencing immune memory and vaccine efficacy.
Collapse
|
11
|
Liu SW, Katsafanas GC, Liu R, Wyatt LS, Moss B. Poxvirus decapping enzymes enhance virulence by preventing the accumulation of dsRNA and the induction of innate antiviral responses. Cell Host Microbe 2015; 17:320-331. [PMID: 25766293 DOI: 10.1016/j.chom.2015.02.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 12/23/2014] [Accepted: 01/28/2015] [Indexed: 11/18/2022]
Abstract
Poxvirus replication involves synthesis of double-stranded RNA (dsRNA), which can trigger antiviral responses by inducing phosphorylation-mediated activation of protein kinase R (PKR) and stimulating 2'5'-oligoadenylate synthetase (OAS). PKR inactivates the translation initiation factor eIF2α via phosphorylation, while OAS induces the endonuclease RNase L to degrade RNA. We show that poxvirus decapping enzymes D9 and D10, which remove caps from mRNAs, inhibit these antiviral responses by preventing dsRNA accumulation. Catalytic site mutations of D9 and D10, but not of either enzyme alone, halt vaccinia virus late protein synthesis and inhibit virus replication. Infection with the D9-D10 mutant was accompanied by massive mRNA reduction, cleavage of ribosomal RNA, and phosphorylation of PKR and eIF2α that correlated with a ∼ 15-fold increase in dsRNA compared to wild-type virus. Additionally, mouse studies show extreme attenuation of the mutant virus. Thus, vaccinia virus decapping, in addition to targeting mRNAs for degradation, prevents dsRNA accumulation and anti-viral responses.
Collapse
Affiliation(s)
- Shin-Wu Liu
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892-3210, USA
| | - George C Katsafanas
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892-3210, USA
| | - Ruikang Liu
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892-3210, USA
| | - Linda S Wyatt
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892-3210, USA
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892-3210, USA.
| |
Collapse
|
12
|
Bidgood SR, Mercer J. Cloak and Dagger: Alternative Immune Evasion and Modulation Strategies of Poxviruses. Viruses 2015; 7:4800-25. [PMID: 26308043 PMCID: PMC4576205 DOI: 10.3390/v7082844] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 12/20/2022] Open
Abstract
As all viruses rely on cellular factors throughout their replication cycle, to be successful they must evolve strategies to evade and/or manipulate the defence mechanisms employed by the host cell. In addition to their expression of a wide array of host modulatory factors, several recent studies have suggested that poxviruses may have evolved unique mechanisms to shunt or evade host detection. These potential mechanisms include mimicry of apoptotic bodies by mature virions (MVs), the use of viral sub-structures termed lateral bodies for the packaging and delivery of host modulators, and the formation of a second, “cloaked” form of infectious extracellular virus (EVs). Here we discuss these various strategies and how they may facilitate poxvirus immune evasion. Finally we propose a model for the exploitation of the cellular exosome pathway for the formation of EVs.
Collapse
Affiliation(s)
- Susanna R Bidgood
- Medical Research Council-Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK.
| | - Jason Mercer
- Medical Research Council-Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
13
|
Ren H, Ferguson BJ, Maluquer de Motes C, Sumner RP, Harman LER, Smith GL. Enhancement of CD8(+) T-cell memory by removal of a vaccinia virus nuclear factor-κB inhibitor. Immunology 2015; 145:34-49. [PMID: 25382035 PMCID: PMC4405322 DOI: 10.1111/imm.12422] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 10/30/2014] [Accepted: 11/03/2014] [Indexed: 12/20/2022] Open
Abstract
Factors influencing T-cell responses are important for vaccine development but are incompletely understood. Here, vaccinia virus (VACV) protein N1 is shown to impair the development of both effector and memory CD8+ T cells and this correlates with its inhibition of nuclear factor-κB (NF-κB) activation. Infection with VACVs that either have the N1L gene deleted (vΔN1) or contain a I6E mutation (vN1.I6E) that abrogates its inhibition of NF-κB resulted in increased central and memory CD8+ T-cell populations, increased CD8+ T-cell cytotoxicity and lower virus titres after challenge. Furthermore, CD8+ memory T-cell function was increased following infection with vN1.I6E, with more interferon-γ production and greater protection against VACV infection following passive transfer to naive mice, compared with CD8+ T cells from mice infected with wild-type virus (vN1.WT). This demonstrates the importance of NF-κB activation within infected cells for long-term CD8+ T-cell memory and vaccine efficacy. Further, it provides a rationale for deleting N1 from VACV vectors to enhance CD8+ T-cell immunogenicity, while simultaneously reducing virulence to improve vaccine safety.
Collapse
Affiliation(s)
- Hongwei Ren
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | | | | | | | | |
Collapse
|
14
|
Smee DF, Dagley A, Downs B, Hagloch J, Tarbet EB. Enhanced efficacy of cidofovir combined with vaccinia immune globulin in treating progressive cutaneous vaccinia virus infections in immunosuppressed hairless mice. Antimicrob Agents Chemother 2015; 59:520-6. [PMID: 25385098 PMCID: PMC4291394 DOI: 10.1128/aac.04289-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 10/31/2014] [Indexed: 12/15/2022] Open
Abstract
The treatment of progressive vaccinia in individuals has involved antiviral drugs, such as cidofovir (CDV), brincidofovir, and/or tecovirimat, combined with vaccinia immune globulin (VIG). VIG is costly, and its supply is limited, so sparing the use of VIG during treatment is an important objective. VIG sparing was modeled in immunosuppressed mice by maximizing the treatment benefits of CDV combined with VIG to determine the effective treatments that delayed the time to death, reduced cutaneous lesion severity, and/or decreased tissue viral titers. SKH-1 hairless mice immunosuppressed with cyclophosphamide and hairless SCID mice (SHO strain) were infected cutaneously with vaccinia virus. Monotherapy, dual combinations (CDV plus VIG), or triple therapy (topical CDV, parenteral CDV, and VIG) were initiated 2 days postinfection and were given every 3 to 4 days through day 11. The efficacy assessment included survival rate, cutaneous lesion severity, and viral titers. Delays in the time to death and the reduction in lesion severity occurred in the following order of efficacy: triple therapy had greater efficacy than double combinations (CDV plus VIG or topical plus parenteral CDV), which had greater efficacy than VIG alone. Parenteral administration of CDV or VIG was necessary to suppress virus titers in internal organs (liver, lung, and spleen). The skin viral titers were significantly reduced by triple therapy only. The greatest efficacy was achieved by triple therapy. In humans, this regimen should translate to a faster cure rate, thus sparing the amount of VIG used for treatment.
Collapse
Affiliation(s)
- Donald F Smee
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - Ashley Dagley
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - Brittney Downs
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - Joseph Hagloch
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - E Bart Tarbet
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, USA
| |
Collapse
|
15
|
Americo JL, Sood CL, Cotter CA, Vogel JL, Kristie TM, Moss B, Earl PL. Susceptibility of the wild-derived inbred CAST/Ei mouse to infection by orthopoxviruses analyzed by live bioluminescence imaging. Virology 2013; 449:120-32. [PMID: 24418545 PMCID: PMC3902144 DOI: 10.1016/j.virol.2013.11.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 10/28/2013] [Accepted: 11/07/2013] [Indexed: 11/25/2022]
Abstract
Classical inbred mice are extensively used for virus research. However, we recently found that some wild-derived inbred mouse strains are more susceptible than classical strains to monkeypox virus. Experiments described here indicated that the 50% lethal dose of vaccinia virus (VACV) and cowpox virus (CPXV) were two logs lower in wild-derived inbred CAST/Ei mice than classical inbred BALB/c mice, whereas there was little difference in the susceptibility of the mouse strains to herpes simplex virus. Live bioluminescence imaging was used to follow spread of pathogenic and attenuated VACV strains and CPXV virus from nasal passages to organs in the chest and abdomen of CAST/Ei mice. Luminescence increased first in the head and then simultaneously in the chest and abdomen in a dose-dependent manner. The spreading kinetics was more rapid with VACV than CPXV although the peak photon flux was similar. These data suggest advantages of CAST/Ei mice for orthopoxvirus studies.
Collapse
Affiliation(s)
- Jeffrey L Americo
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cindy L Sood
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Catherine A Cotter
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jodi L Vogel
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas M Kristie
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Patricia L Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Sumner RP, Ren H, Smith GL. Deletion of immunomodulator C6 from vaccinia virus strain Western Reserve enhances virus immunogenicity and vaccine efficacy. J Gen Virol 2013; 94:1121-1126. [PMID: 23288427 PMCID: PMC3709586 DOI: 10.1099/vir.0.049700-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 01/03/2013] [Indexed: 01/18/2023] Open
Abstract
Vectors based on vaccinia virus (VACV), the vaccine used to eradicate smallpox, are currently popular candidates for the vaccination against numerous infectious diseases including malaria and AIDS. Although VACV induces robust cellular and humoral responses, enhancing the safety and efficacy of these vectors remains an important area of research. Here, we describe the enhanced immunogenicity of a recombinant VACV Western Reserve (WR) strain lacking the immunomodulatory protein C6 (vΔC6). Intradermal infection of mice with vΔC6 was shown previously to induce smaller lesions, indicating viral attenuation, and this was confirmed here using a different inoculation dose. In addition, data presented show that vaccination with vΔC6 provided better protection against challenge with a lethal dose of VACV WR, indicating this virus is a better vaccine. Increased protection was not due to improved humoral responses, but instead enhanced cytotoxic activity of T-cells 1 month post-inoculation in the spleens of vΔC6-vaccinated mice.
Collapse
Affiliation(s)
- Rebecca P Sumner
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Hongwei Ren
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Geoffrey L Smith
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
17
|
He Y, Wang Y, Struble EB, Zhang P, Chowdhury S, Reed JL, Kennedy M, Scott DE, Fisher RW. Epitope mapping by random peptide phage display reveals essential residues for vaccinia extracellular enveloped virion spread. Virol J 2012; 9:217. [PMID: 23006741 PMCID: PMC3495767 DOI: 10.1186/1743-422x-9-217] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 09/14/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A33 is a type II integral membrane protein expressed on the extracellular enveloped form of vaccinia virus (VACV). Passive transfer of A33-directed monoclonal antibodies or vaccination with an A33 subunit vaccine confers protection against lethal poxvirus challenge in animal models. Homologs of A33 are highly conserved among members of the Orthopoxvirus genus and are potential candidates for inclusion in vaccines or assays targeting extracellular enveloped virus activity. One monoclonal antibody directed against VACV A33, MAb-1G10, has been shown to target a conformation-dependent epitope. Interestingly, while it recognizes VACV A33 as well as the corresponding variola homolog, it does not bind to the monkeypox homolog. In this study, we utilized a random phage display library to investigate the epitope recognized by MAb-1G10 that is critical for facilitating cell-to-cell spread of the vaccinia virus. RESULTS By screening with linear or conformational random phage libraries, we found that phages binding to MAb-1G10 display the consensus motif CEPLC, with a disulfide bond formed between two cysteine residues required for MAb-1G10 binding. Although the phage motif contained no linear sequences homologous to VACV A33, structure modeling and analysis suggested that residue D115 is important to form the minimal epitope core. A panel of point mutants expressing the ectodomain of A33 protein was generated and analyzed by either binding assays such as ELISA and immunoprecipitation or a functional assessment by blocking MAb-1G10 mediated comet inhibition in cell culture. CONCLUSIONS These results confirm L118 as a component of the MAb-1G10 binding epitope, and further identify D115 as an essential residue. By defining the minimum conformational structure, as well as the conformational arrangement of a short peptide sequence recognized by MAb-1G10, these results introduce the possibility of designing small molecule mimetics that may interfere with the function of A33 in vivo. This information will also be useful for designing improved assays to evaluate the potency of monoclonal and polyclonal products that target A33 or A33-modulated EV dissemination.
Collapse
Affiliation(s)
- Yong He
- Laboratory of Plasma Derivatives, Division of Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, FDA/CBER/OBRR/DH/LPD, HFM-345, 1401 Rockville Pike, Rockville, MD 20852, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Memory CD8+ T cells specific for a single immunodominant or subdominant determinant induced by peptide-dendritic cell immunization protect from an acute lethal viral disease. J Virol 2012; 86:9748-59. [PMID: 22740418 DOI: 10.1128/jvi.00981-12] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The antigens recognized by individual CD8(+) T cells are small peptides bound to major histocompatibility complex (MHC) class I molecules. The CD8(+) T cell response to a virus is restricted to several peptides, and the magnitudes of the effector as well as memory phases of the response to the individual peptides are generally hierarchical. The peptide eliciting a stronger response is called immunodominant (ID), and those with smaller-magnitude responses are termed subdominant (SD). The relative importance of ID and SD determinants in protective immunity remains to be fully elucidated. We previously showed that multispecific memory CD8(+) T cells can protect susceptible mice from mousepox, an acute lethal viral disease. It remained unknown, however, whether CD8(+) T cells specific for single ID or SD peptides could be protective. Here, we demonstrate that immunization with dendritic cells pulsed with ID and some but not all SD peptides induces memory CD8(+) T cells that are fully capable of protecting susceptible mice from mousepox. Additionally, while natural killer (NK) cells are essential for the natural resistance of nonimmune C57BL/6 (B6) to mousepox, we show that memory CD8(+) T cells of single specificity also protect B6 mice depleted of NK cells. This suggests it is feasible to produce effective antiviral CD8(+) T cell vaccines using single CD8(+) T cell determinants and that NK cells are no longer essential when memory CD8(+) T cells are present.
Collapse
|
19
|
Johnson BF, Kanatani Y, Fujii T, Saito T, Yokote H, Smith GL. Serological responses in humans to the smallpox vaccine LC16m8. J Gen Virol 2011; 92:2405-2410. [PMID: 21715598 PMCID: PMC3347799 DOI: 10.1099/vir.0.034207-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 06/28/2011] [Indexed: 11/18/2022] Open
Abstract
In response to potential bioterrorism with smallpox, members of the Japanese Self-Defense Forces were vaccinated with vaccinia virus (VACV) strain LC16m8, an attenuated smallpox vaccine derived from VACV strain Lister. The serological response induced by LC16m8 to four virion-surface proteins and the intracellular mature virus (IMV) and extracellular enveloped virus (EEV) was investigated. LC16m8 induced antibody response against the IMV protein A27 and the EEV protein A56. LC16m8 also induced IMV-neutralizing antibodies, but unlike the VACV strain Lister, did not induce either EEV-neutralizing antibody or antibody to EEV protein B5, except after revaccination. Given that B5 is the only target for EEV-neutralizing antibody and that neutralization of both IMV and EEV give optimal protection against orthopoxvirus challenge, these data suggest that immunity induced by LC16m8 might be less potent than that deriving from strain Lister. This potential disadvantage should be balanced against the advantage of the greater safety of LC16m8.
Collapse
Affiliation(s)
- Benjamin F. Johnson
- Section of Virology, Faculty of Medicine, Imperial College London, St Mary’s Campus, Norfolk Place, London W2 1PG, UK
| | - Yasuhiro Kanatani
- National Institute of Public Health, Department of Policy Sciences, 2-3-6 Minami, Wako-shi, Saitama 351-0197, Japan
| | - Tatsuya Fujii
- Self-Defense Force, Central Hospital, Health Department, Management Division, 1-2-24 Ikejiri, Setagaya, Tokyo 154-8532, Japan
| | - Tomoya Saito
- Bio-preparedness Research Laboratory, Department of Tropical Medicine and Parasitology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Hiroyuki Yokote
- The Chemo-Sero-Therapeutic Research Institute (KAKETSUKEN), 1-6-1 Okubo, Kumamoto 860-8568, Japan
| | - Geoffrey L. Smith
- Section of Virology, Faculty of Medicine, Imperial College London, St Mary’s Campus, Norfolk Place, London W2 1PG, UK
| |
Collapse
|
20
|
Zaitseva M, Kapnick SM, Meseda CA, Shotwell E, King LR, Manischewitz J, Scott J, Kodihalli S, Merchlinsky M, Nielsen H, Lantto J, Weir JP, Golding H. Passive immunotherapies protect WRvFire and IHD-J-Luc vaccinia virus-infected mice from lethality by reducing viral loads in the upper respiratory tract and internal organs. J Virol 2011; 85:9147-58. [PMID: 21715493 PMCID: PMC3165812 DOI: 10.1128/jvi.00121-11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 06/13/2011] [Indexed: 02/04/2023] Open
Abstract
Whole-body bioimaging was employed to study the effects of passive immunotherapies on lethality and viral dissemination in BALB/c mice challenged with recombinant vaccinia viruses expressing luciferase. WRvFire and IHD-J-Luc vaccinia viruses induced lethality with similar times to death following intranasal infection, but WRvFire replicated at higher levels than IHD-J-Luc in the upper and lower respiratory tracts. Three types of therapies were tested: licensed human anti-vaccinia virus immunoglobulin intravenous (VIGIV); recombinant anti-vaccinia virus immunoglobulin (rVIG; Symphogen, Denmark), an investigational product containing a mixture of 26 human monoclonal antibodies (HuMAbs) against mature virion (MV) and enveloped virion (EV); and HuMAb compositions targeting subsets of MV or EV proteins. Bioluminescence recorded daily showed that pretreatment with VIGIV (30 mg) or with rVIG (100 μg) on day -2 protected mice from death but did not prevent viral replication at the site of inoculation and dissemination to internal organs. Compositions containing HuMAbs against MV or EV proteins were protective in both infection models at 100 μg per animal, but at 30 μg, only anti-EV antibodies conferred protection. Importantly, the t statistic of the mean total fluxes revealed that viral loads in surviving mice were significantly reduced in at least 3 sites for 3 consecutive days (days 3 to 5) postchallenge, while significant reduction for 1 or 2 days in any individual site did not confer protection. Our data suggest that reduction of viral replication at multiple sites, including respiratory tract, spleen, and liver, as monitored by whole-body bioluminescence can be used to predict the effectiveness of passive immunotherapies in mouse models.
Collapse
Affiliation(s)
- Marina Zaitseva
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Building 29B, Room 4NN06, 8800 Rockville Pike, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Xu C, Meng X, Yan B, Crotty S, Deng J, Xiang Y. An epitope conserved in orthopoxvirus A13 envelope protein is the target of neutralizing and protective antibodies. Virology 2011; 418:67-73. [PMID: 21810533 PMCID: PMC3163717 DOI: 10.1016/j.virol.2011.06.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 06/01/2011] [Accepted: 06/17/2011] [Indexed: 11/27/2022]
Abstract
Primary immunization of humans with smallpox vaccine (live vaccinia virus (VACV)) consistently elicits antibody responses to six VACV virion membrane proteins, including A13. However, whether anti-A13 antibody contributes to immune protection against orthopoxviruses was unknown. Here, we isolated a murine monoclonal antibody (mAb) against A13 from a mouse that had been infected with VACV. The anti-A13 mAb bound to recombinant A13 protein with an affinity of 3.4 nM and neutralized VACV mature virions. Passive immunization of mice with the anti-A13 mAb protected against intranasal VACV infection. The epitope of the anti-A13 mAb was mapped to a 10-amino acid sequence conserved in all orthopoxviruses, including viriola virus and monkeypox virus, suggesting that anti-A13 antibodies elicited by smallpox vaccine might contribute to immune protection against orthopoxviruses. In addition, our data demonstrates that anti-A13 mAbs are effective for treating orthopoxvirus infection.
Collapse
Affiliation(s)
- Chungui Xu
- Department of Microbiology and Immunology, Univ. of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | | | | | | | | |
Collapse
|
22
|
Berhanu A, King DS, Mosier S, Jordan R, Jones KF, Hruby DE, Grosenbach DW. Impact of ST-246® on ACAM2000™ smallpox vaccine reactogenicity, immunogenicity, and protective efficacy in immunodeficient mice. Vaccine 2010; 29:289-303. [PMID: 21036130 PMCID: PMC3023305 DOI: 10.1016/j.vaccine.2010.10.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 09/29/2010] [Accepted: 10/13/2010] [Indexed: 11/16/2022]
Abstract
Although a highly effective vaccine against smallpox, vaccinia virus (VV) is not without adverse events, some of which can be life-threatening, particularly in immunocompromised individuals. We have recently demonstrated that the immunogenicity and protective efficacy of Dryvax(®) in immunocompetent mice is preserved even when co-administered with ST-246, an orally bioavailable small-molecule inhibitor of orthopoxvirus egress and dissemination. In addition, ST-246 markedly reduced the reactogenicity of the smallpox vaccine ACAM2000 and the highly neurovirulent VV strain Western Reserve (VV-WR). Here, we evaluated the impact of ST-246 co-administration on ACAM2000 reactogenicity, immunogenicity, and protective efficacy in seven murine models of varying degrees of humoral and cellular immunodeficiency: BALB/c and B-cell deficient (JH-KO) mice depleted of CD4(+) or CD8(+) or both subsets of T cells. We observed that ST-246 reduced vaccine lesion severity and time to complete resolution in all of the immunodeficient models examined, except in those lacking both CD4(+) and CD8(+) T cells. Although VV-specific humoral responses were moderately reduced by ST-246 treatment, cellular responses were generally comparable or slightly enhanced at both 1 and 6 months post-vaccination. Most importantly, in those models in which vaccination given alone conferred protection against lethal VV challenge, similar levels of protection were observed at both time points when vaccination was given with ST-246. These data suggest that, with the exception of individuals with irreversible, combined CD4(+) and CD8(+) T-cell deficiency, ST-246 co-administered at the time of vaccination may help reduce vaccine reactogenicity--even in those lacking humoral immunity--without impeding the induction of protective immunity.
Collapse
Affiliation(s)
- Aklile Berhanu
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | - David S. King
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | - Stacie Mosier
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | - Robert Jordan
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | - Kevin F. Jones
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | - Dennis E. Hruby
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | | |
Collapse
|
23
|
Postexposure prevention of progressive vaccinia in SCID mice treated with vaccinia immune globulin. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 18:67-74. [PMID: 21106779 DOI: 10.1128/cvi.00280-10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A recently reported case of progressive vaccinia (PV) in an immunocompromised patient has refocused attention on this condition. Uniformly fatal prior to the licensure of vaccinia immune globulin (VIG) in 1978, PV was still fatal in about half of VIG-treated patients overall, with a greater mortality rate in infants and children. Additional therapies would be needed in the setting of a smallpox bioterror event, since mass vaccination following any variola virus release would inevitably result in exposure of immunocompromised people through vaccination or contact with vaccinees. Well-characterized animal models of disease can support the licensure of new products when human studies are not ethical or feasible, as in the case of PV. We chose vaccinia virus-scarified SCID mice to model PV. As in immunocompromised humans, vaccinia virus-scarified SCID animals develop enlarging primary lesions with minimal or no inflammation, eventual distal virus spread, and lethal outcomes if left untreated. Postexposure treatment with VIG slowed disease progression, caused local lesion regression, and resulted in the healthy survival of most of the mice for more than 120 days. Combination treatment with VIG and topical cidofovir also resulted in long-term disease-free survival of most of the animals, even when initiated 7 days postinfection. These results support the possibility that combination treatments may be effective in humans and support using this SCID model of PV to test new antibody therapies and combination therapies and to provide further insights into the pathogenesis and treatment of PV.
Collapse
|
24
|
Xiao Y, Isaacs SN. Therapeutic Vaccines and Antibodies for Treatment of Orthopoxvirus Infections. Viruses 2010; 2:2381-2403. [PMID: 21197387 PMCID: PMC3011862 DOI: 10.3390/v2102381] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2010] [Revised: 10/09/2010] [Accepted: 10/13/2010] [Indexed: 11/16/2022] Open
Abstract
Despite the eradication of smallpox several decades ago, variola and monkeypox viruses still have the potential to become significant threats to public health. The current licensed live vaccinia virus-based smallpox vaccine is extremely effective as a prophylactic vaccine to prevent orthopoxvirus infections, but because of safety issues, it is no longer given as a routine vaccine to the general population. In the event of serious human orthopoxvirus infections, it is important to have treatments available for individual patients as well as their close contacts. The smallpox vaccine and vaccinia immune globulin (VIG) were used in the past as therapeutics for patients exposed to smallpox. VIG was also used in patients who were at high risk of developing complications from smallpox vaccination. Thus post-exposure vaccination and VIG treatments may again become important therapeutic modalities. This paper summarizes some of the historic use of the smallpox vaccine and immunoglobulins in the post-exposure setting in humans and reviews in detail the newer animal studies that address the use of therapeutic vaccines and immunoglobulins in orthopoxvirus infections as well as the development of new therapeutic monoclonal antibodies.
Collapse
Affiliation(s)
- Yuhong Xiao
- Division of Infectious Diseases, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA; E-Mail:
| | - Stuart N. Isaacs
- Division of Infectious Diseases, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA; E-Mail:
- Infectious Diseases Section, Philadelphia Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| |
Collapse
|
25
|
Breiman A, Smith GL. Vaccinia virus B5 protein affects the glycosylation, localization and stability of the A34 protein. J Gen Virol 2010; 91:1823-7. [PMID: 20200189 PMCID: PMC3052527 DOI: 10.1099/vir.0.020677-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 03/02/2010] [Indexed: 11/18/2022] Open
Abstract
Vaccinia virus has two infectious forms, the intracellular mature virus, which has a single envelope, and the extracellular enveloped virus (EEV), which is surrounded by two lipid bilayers. The outer membrane of the EEV contains at least six viral proteins. Among them A34, a type II membrane glycoprotein, and B5, a type I membrane glycoprotein, form a complex and are involved in processes such as morphogenesis and EEV entry. A34 is required for normal incorporation of B5 into the EEV membrane. Here, we used a virus lacking B5 and viruses with mutations in the B5 membrane-proximal stalk region and looked at the effect of those modifications on A34. Data presented show that B5 is required for the correct glycosylation, trafficking and stability of A34, emphasizing the complex interactions and mutual dependence of these vaccinia EEV proteins.
Collapse
Affiliation(s)
- Adrien Breiman
- Department of Virology, Faculty of Medicine, Imperial College London, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| | | |
Collapse
|
26
|
von Krempelhuber A, Vollmar J, Pokorny R, Rapp P, Wulff N, Petzold B, Handley A, Mateo L, Siersbol H, Kollaritsch H, Chaplin P. A randomized, double-blind, dose-finding Phase II study to evaluate immunogenicity and safety of the third generation smallpox vaccine candidate IMVAMUNE. Vaccine 2010; 28:1209-16. [PMID: 19944151 PMCID: PMC2814951 DOI: 10.1016/j.vaccine.2009.11.030] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Revised: 11/04/2009] [Accepted: 11/11/2009] [Indexed: 01/27/2023]
Abstract
IMVAMUNE is a Modified Vaccinia Ankara (MVA)-based virus that is being developed as a safer 3rd generation smallpox vaccine. In order to determine the optimal dose for further development, a double-blind, randomized Phase II trial was performed testing three different doses of IMVAMUNE in 164 healthy volunteers. All three IMVAMUNE doses displayed a favourable safety profile, with local reactions as the most frequent observation. The 1 x 10(8)TCID(50) IMVAMUNE dose induced a total antibody response in 94% of the subjects following the first vaccination and the highest peak seroconversion rates by ELISA (100%) and PRNT (71%). This IMVAMUNE dose was considered to be optimal for the further clinical development of this highly attenuated poxvirus as a safer smallpox vaccine.
Collapse
|
27
|
Benhnia MREI, McCausland MM, Laudenslager J, Granger SW, Rickert S, Koriazova L, Tahara T, Kubo RT, Kato S, Crotty S. Heavily isotype-dependent protective activities of human antibodies against vaccinia virus extracellular virion antigen B5. J Virol 2009; 83:12355-67. [PMID: 19793826 PMCID: PMC2786738 DOI: 10.1128/jvi.01593-09] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Accepted: 09/17/2009] [Indexed: 11/20/2022] Open
Abstract
Antibodies against the extracellular virion (EV or EEV) form of vaccinia virus are an important component of protective immunity in animal models and likely contribute to the protection of immunized humans against poxviruses. Using fully human monoclonal antibodies (MAbs), we now have shown that the protective attributes of the human anti-B5 antibody response to the smallpox vaccine (vaccinia virus) are heavily dependent on effector functions. By switching Fc domains of a single MAb, we have definitively shown that neutralization in vitro--and protection in vivo in a mouse model--by the human anti-B5 immunoglobulin G MAbs is isotype dependent, thereby demonstrating that efficient protection by these antibodies is not simply dependent on binding an appropriate vaccinia virion antigen with high affinity but in fact requires antibody effector function. The complement components C3 and C1q, but not C5, were required for neutralization. We also have demonstrated that human MAbs against B5 can potently direct complement-dependent cytotoxicity of vaccinia virus-infected cells. Each of these results was then extended to the polyclonal human antibody response to the smallpox vaccine. A model is proposed to explain the mechanism of EV neutralization. Altogether these findings enhance our understanding of the central protective activities of smallpox vaccine-elicited antibodies in immunized humans.
Collapse
Affiliation(s)
- Mohammed Rafii-El-Idrissi Benhnia
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California 92037, Kyowa Hakko Kirin California, La Jolla, California 92037
| | - Megan M. McCausland
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California 92037, Kyowa Hakko Kirin California, La Jolla, California 92037
| | - John Laudenslager
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California 92037, Kyowa Hakko Kirin California, La Jolla, California 92037
| | - Steven W. Granger
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California 92037, Kyowa Hakko Kirin California, La Jolla, California 92037
| | - Sandra Rickert
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California 92037, Kyowa Hakko Kirin California, La Jolla, California 92037
| | - Lilia Koriazova
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California 92037, Kyowa Hakko Kirin California, La Jolla, California 92037
| | - Tomoyuki Tahara
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California 92037, Kyowa Hakko Kirin California, La Jolla, California 92037
| | - Ralph T. Kubo
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California 92037, Kyowa Hakko Kirin California, La Jolla, California 92037
| | - Shinichiro Kato
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California 92037, Kyowa Hakko Kirin California, La Jolla, California 92037
| | - Shane Crotty
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, California 92037, Kyowa Hakko Kirin California, La Jolla, California 92037
| |
Collapse
|
28
|
The smallpox vaccine induces an early neutralizing IgM response. Vaccine 2009; 28:140-7. [PMID: 19822230 DOI: 10.1016/j.vaccine.2009.09.086] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 08/25/2009] [Accepted: 09/23/2009] [Indexed: 12/12/2022]
Abstract
The antibody response elicited after immunization with vaccinia virus (VacV) is known to be sufficient to confer host protection against VacV or smallpox. In humans it has been shown that such anti-VacV antibody production can be sustained for decades. Nevertheless, little is known about the kinetics and the role in protection of the early antibody response after vaccination. In this study we identify VacV neutralizing IgM antibodies as early as 4 days after infection of C57BL/6 mice. Most of this IgM production is T cell dependent and predominantly independent of the germinal center reaction (SAP/SH2D1A independent). Importantly, the IgM neutralized both infectious forms of VacV: the intracellular mature virion (MV, IMV) and the extracellular enveloped virion (EV, EEV). Moreover, in mice primed with MHCII restricted peptides, an increase in the total VacV neutralizing antibody titers was seen, a large component of which was neutralizing IgM against the same protein from which the priming peptide was derived. To further demonstrate the biological relevance of this early neutralizing response, we examined anti-VacV antibodies in humans after vaccination. Human subjects could be divided into two groups early after immunization: IgG(hi) and IgG(lo). VacV IgM neutralizing antibodies were detected in the IgG(lo) group. Taken together these results indicate that both in a small animal model and in humans an early neutralizing IgM response after VacV immunization is present and likely contributes to control of the infection prior to the development of a robust IgG response.
Collapse
|
29
|
Shinoda K, Wyatt LS, Irvine KR, Moss B. Engineering the vaccinia virus L1 protein for increased neutralizing antibody response after DNA immunization. Virol J 2009; 6:28. [PMID: 19257896 PMCID: PMC2654435 DOI: 10.1186/1743-422x-6-28] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Accepted: 03/03/2009] [Indexed: 12/22/2022] Open
Abstract
Background The licensed smallpox vaccine, comprised of infectious vaccinia virus, has associated adverse effects, particularly for immunocompromised individuals. Therefore, safer DNA and protein vaccines are being investigated. The L1 protein, a component of the mature virion membrane that is conserved in all sequenced poxviruses, is required for vaccinia virus entry into host cells and is a target for neutralizing antibody. When expressed by vaccinia virus, the unglycosylated, myristoylated L1 protein attaches to the viral membrane via a C-terminal transmembrane anchor without traversing the secretory pathway. The purpose of the present study was to investigate modifications of the gene expressing the L1 protein that would increase immunogenicity in mice when delivered by a gene gun. Results The L1 gene was codon modified for optimal expression in mammalian cells and potential N-glycosylation sites removed. Addition of a signal sequence to the N-terminus of L1 increased cell surface expression as shown by confocal microscopy and flow cytometry of transfected cells. Removal of the transmembrane domain led to secretion of L1 into the medium. Induction of binding and neutralizing antibodies in mice was enhanced by gene gun delivery of L1 containing the signal sequence with or without the transmembrane domain. Each L1 construct partially protected mice against weight loss caused by intranasal administration of vaccinia virus. Conclusion Modifications of the vaccinia virus L1 gene including codon optimization and addition of a signal sequence with or without deletion of the transmembrane domain can enhance the neutralizing antibody response of a DNA vaccine.
Collapse
Affiliation(s)
- Kaori Shinoda
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892-3210, USA.
| | | | | | | |
Collapse
|
30
|
Lustig S, Maik-Rachline G, Paran N, Melamed S, Israely T, Erez N, Orr N, Reuveny S, Ordentlich A, Laub O, Shafferman A, Velan B. Effective post-exposure protection against lethal orthopoxviruses infection by vaccinia immune globulin involves induction of adaptive immune response. Vaccine 2009; 27:1691-9. [PMID: 19195492 DOI: 10.1016/j.vaccine.2009.01.038] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 12/16/2008] [Accepted: 01/15/2009] [Indexed: 11/28/2022]
Abstract
The therapeutic potential of human vaccinia immunoglobulin (VIG) in orthopoxvirus infection was examined using two mouse models for human poxvirus, based on Ectromelia virus and Vaccinia Western Reserve (WR) respiratory infections. Despite the relatively fast clearance of human VIG from mice circulation, a single VIG injection protected immune-competent mice against both infections. Full protection against lethal Ectromelia virus infection was achieved by VIG injection up to one day post-exposure, and even injection of VIG two or three days post-infection conferred solid protection (60-80%). Nevertheless, VIG failed to protect VACV-WR challenged immune-deficient mice, even though repeated injections prolonged SCID mice survival. These results suggest the involvement of host immunity in protection. VIG provides the initial protective time-window allowing induction of the adaptive response required to achieve complete protection. Additionally, VIG can be administered in conjunction with active Vaccinia-Lister vaccination. Vaccine efficiency is not impaired, providing a non-prohibitive VIG dose is used. Thus, VIG can be used as a prophylactic measure against post-vaccinal complications but could also serve for post-exposure treatment against smallpox.
Collapse
Affiliation(s)
- Shlomo Lustig
- Department of Infectious Diseases, Israel Institute for Biological Research, Israel.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Midgley CM, Putz MM, Weber JN, Smith GL. Vaccinia virus strain NYVAC induces substantially lower and qualitatively different human antibody responses compared with strains Lister and Dryvax. J Gen Virol 2009; 89:2992-2997. [PMID: 19008384 PMCID: PMC2885029 DOI: 10.1099/vir.0.2008/004440-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The antibody responses elicited by immunization of humans with vaccinia virus (VACV) strains Lister, Dryvax and NYVAC have been determined and compared. Neutralizing antibodies against intracellular mature virus (IMV) and extracellular enveloped virus (EEV), and binding antibody titres (ELISA) against the EEV protein B5, the IMV proteins A27 and H3, and VACV-infected cell lysate were measured. Lister and Dryvax induced broadly similar antibody titres, consistent with the fact that these vaccines each protected against smallpox. In contrast, antibody titres induced by NYVAC were significantly lower than those induced by both Lister and Dryvax. Moreover, there were qualitative differences with NYVAC-immunized subjects failing to induce A27-specific antibodies. These observations suggest that although NYVAC is a safer VACV strain, it does not induce an optimal VACV-specific antibody response. However, NYVAC strains engineered to express antigens from other pathogens remain promising candidate vaccines for immunization against other diseases.
Collapse
Affiliation(s)
- Claire M Midgley
- Department of Virology, Faculty of Medicine, Imperial College London, Norfolk Place, London W2 1PG, UK
| | - Mike M Putz
- Department of Virology, Faculty of Medicine, Imperial College London, Norfolk Place, London W2 1PG, UK
| | - Jonathan N Weber
- Division of Medicine, Imperial College London, Norfolk Place, London W2 1PG, UK
| | - Geoffrey L Smith
- Department of Virology, Faculty of Medicine, Imperial College London, Norfolk Place, London W2 1PG, UK
| |
Collapse
|
32
|
Vaccinia virus extracellular enveloped virion neutralization in vitro and protection in vivo depend on complement. J Virol 2008; 83:1201-15. [PMID: 19019965 DOI: 10.1128/jvi.01797-08] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antibody neutralization is an important component of protective immunity against vaccinia virus (VACV). Two distinct virion forms, mature virion and enveloped virion (MV and EV, respectively), possess separate functions and nonoverlapping immunological properties. In this study we examined the mechanics of EV neutralization, focusing on EV protein B5 (also called B5R). We show that neutralization of EV is predominantly complement dependent. From a panel of high-affinity anti-B5 monoclonal antibodies (MAbs), the only potent neutralizer in vitro (90% at 535 ng/ml) was an immunoglobulin G2a (IgG2a), and neutralization was complement mediated. This MAb was the most protective in vivo against lethal intranasal VACV challenge. Further studies demonstrated that in vivo depletion of complement caused a >50% loss of anti-B5 IgG2a protection, directly establishing the importance of complement for protection against the EV form. However, the mechanism of protection is not sterilizing immunity via elimination of the inoculum as the viral inoculum consisted of a purified MV form. The prevention of illness in vivo indicated rapid control of infection. We further demonstrate that antibody-mediated killing of VACV-infected cells expressing surface B5 is a second protective mechanism provided by complement-fixing anti-B5 IgG. Cell killing was very efficient, and this effector function was highly isotype specific. These results indicate that anti-B5 antibody-directed cell lysis via complement is a powerful mechanism for clearance of infected cells, keeping poxvirus-infected cells from being invisible to humoral immune responses. These findings highlight the importance of multiple mechanisms of antibody-mediated protection against VACV and point to key immunobiological differences between MVs and EVs that impact the outcome of infection.
Collapse
|
33
|
Nelson GE, Sisler JR, Chandran D, Moss B. Vaccinia virus entry/fusion complex subunit A28 is a target of neutralizing and protective antibodies. Virology 2008; 380:394-401. [PMID: 18789472 PMCID: PMC2628551 DOI: 10.1016/j.virol.2008.08.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2008] [Revised: 07/25/2008] [Accepted: 08/05/2008] [Indexed: 11/20/2022]
Abstract
The vaccinia virus entry/fusion complex (EFC) is comprised of at least eight transmembrane proteins that are conserved in all poxviruses. However, neither the physical structure of the EFC nor the immunogenicity of the individual components has been determined. We prepared soluble forms of two EFC components, A28 and H2, by replacing the transmembrane domain with a signal peptide and adding a polyhistidine tail. The proteins were expressed by baculoviruses, secreted from insect cells, purified by affinity chromatography and used to raise antibodies in rabbits. The antibodies recognized the viral proteins but only the antibody to recombinant A28 bound intact virions and neutralized infectivity. Analyses with a set of overlapping peptides revealed a neutralizing epitope between residues 73 and 92 of A28. Passive immunization of mice with IgG purified from the anti-A28 serum provided partial protection against a vaccinia virus intranasal challenge, whereas IgG from the anti-H2 serum did not.
Collapse
Affiliation(s)
- Gretchen E. Nelson
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-3210
| | - Jerry R. Sisler
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-3210
| | | | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-3210
| |
Collapse
|
34
|
Vaccinia virus encodes I5, a small hydrophobic virion membrane protein that enhances replication and virulence in mice. J Virol 2008; 82:10071-8. [PMID: 18701595 DOI: 10.1128/jvi.01355-08] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The vaccinia virus I5L open reading frame encodes a 79-amino-acid protein, with two predicted transmembrane domains, that is conserved among all sequenced members of the chordopoxvirus subfamily. No nonpoxvirus homologs or functional motifs have been recognized, and the role of the I5 protein remains unknown. We found that synthesis of I5 was dependent on viral DNA replication and occurred exclusively at late times, consistent with a consensus late promoter motif adjacent to the start of the open reading frame. I5 was present in preparations of purified virions and could be extracted with nonionic detergent, suggesting membrane insertion. Transmission electron microscopy of immunogold-labeled thawed cryosections of infected cells revealed the association of an epitope-tagged I5 with the membranes of immature and mature virions. Viable I5L deletion and frameshift mutants were constructed and found to replicate like wild-type virus in a variety of cell lines and primary human epidermal keratinocytes, indicating that the protein was dispensable for in vitro cultivation. However, mouse intranasal challenge experiments indicated that a mutant virus with a frameshift resulting in a stop codon near the N terminus of I5 was attenuated compared to control virus. The attenuation was correlated with clearance of mutant viruses from the respiratory tract and with less progression and earlier resolution of pathological changes. We suggest that I5 is involved in an aspect of host defense that is evolutionarily conserved although a role in cell tropism should also be considered.
Collapse
|
35
|
Disparity between levels of in vitro neutralization of vaccinia virus by antibody to the A27 protein and protection of mice against intranasal challenge. J Virol 2008; 82:8022-9. [PMID: 18524827 DOI: 10.1128/jvi.00568-08] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Immunization with recombinant proteins may provide a safer alternative to live vaccinia virus for prophylaxis of poxvirus infections. Although antibody protects against vaccinia virus infection, the mechanism is not understood and the selection of immunogens is daunting as there are dozens of surface proteins and two infectious forms known as the mature virion (MV) and the enveloped virion (EV). Our previous studies showed that mice immunized with soluble forms of EV membrane proteins A33 and B5 and MV membrane protein L1 or passively immunized with antibodies to these proteins survived an intranasal challenge with vaccinia virus. The present study compared MV protein A27, which has a role in virus attachment to glycosaminoglycans on the cell surface, to L1 with respect to immunogenicity and protection. Although mice developed similar levels of neutralizing antibody after immunizations with A27 or L1, A27-immunized mice exhibited more severe disease upon an intranasal challenge with vaccinia virus. In addition, mice immunized with A27 and A33 were not as well protected as mice receiving L1 and A33. Polyclonal rabbit anti-A27 and anti-L1 IgG had equivalent MV-neutralizing activities when measured by the prevention of infection of human or mouse cells or cells deficient in glycosaminoglycans or by adding antibody prior to or after virus adsorption. Nevertheless, the passive administration of antibody to A27 was poorly protective compared to the antibody to L1. These studies raise questions regarding the basis for antibody protection against poxvirus disease and highlight the importance of animal models for the early evaluation of vaccine candidates.
Collapse
|
36
|
Senkevich TG, Wyatt LS, Weisberg AS, Koonin EV, Moss B. A conserved poxvirus NlpC/P60 superfamily protein contributes to vaccinia virus virulence in mice but not to replication in cell culture. Virology 2008; 374:506-14. [PMID: 18281072 PMCID: PMC2412903 DOI: 10.1016/j.virol.2008.01.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 01/02/2008] [Accepted: 01/07/2008] [Indexed: 10/22/2022]
Abstract
Of the vaccinia virus genes that are conserved in all sequenced poxviruses, each one except for VACWR084 (G6R) has been at least partially characterized. The poxvirus protein encoded by G6R belongs to the NlpC/P60 superfamily, which consists of proteins with a papain-like fold and known or predicted protease, amidase or acyltransferase activity. The G6 protein was synthesized late in infection and localized to the interior of virions, primarily between the membrane and core. Unlike other conserved poxvirus genes, G6R was not required for virus propagation and spread in a variety of cells. Nevertheless, G6R null mutants caused less severe disease in mice than the parent or revertant virus. Moreover, mutation of the predicted catalytic cysteine led to the same level of attenuation as a null mutant, suggesting that the G6 protein has enzymatic activity that is important in vivo. Conservation of G6R amongst poxviruses and the disparity between its role in vitro and in vivo imply that the protein is involved in an aspect of the virus-host interaction that is common to vertebrates and insects.
Collapse
Affiliation(s)
- Tatiana G. Senkevich
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Linda S. Wyatt
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrea S. Weisberg
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eugene V. Koonin
- National Center for Biotechnology Information, National Library of Medicine, Bethesda, MD 20894
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
37
|
Vaccination of BALB/c mice with Escherichia coli-expressed vaccinia virus proteins A27L, B5R, and D8L protects mice from lethal vaccinia virus challenge. J Virol 2008; 82:3517-29. [PMID: 18199639 DOI: 10.1128/jvi.01854-07] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The potential threat of smallpox use in a bioterrorist attack has heightened the need to develop an effective smallpox vaccine for immunization of the general public. Vaccination with the current smallpox vaccine, Dryvax, produces protective immunity but may result in adverse reactions for some vaccinees. A subunit vaccine composed of protective vaccinia virus proteins should avoid the complications arising from live-virus vaccination and thus provide a safer alternative smallpox vaccine. In this study, we assessed the protective efficacy and immunogenicity of a multisubunit vaccine composed of the A27L and D8L proteins from the intracellular mature virus (IMV) form and the B5R protein from the extracellular enveloped virus (EEV) form of vaccinia virus. BALB/c mice were immunized with Escherichia coli-produced A27L, D8L, and B5R proteins in an adjuvant consisting of monophosphoryl lipid A and trehalose dicorynomycolate or in TiterMax Gold adjuvant. Following immunization, mice were either sacrificed for analysis of immune responses or lethally challenged by intranasal inoculation with vaccinia virus strain Western Reserve. We observed that three immunizations either with A27L, D8L, and B5R or with the A27L and B5R proteins alone induced potent neutralizing antibody responses and provided complete protection against lethal vaccinia virus challenge. Several linear B-cell epitopes within the three proteins were recognized by sera from the immunized mice. In addition, protein-specific cellular responses were detected in spleens of immunized mice by a gamma interferon enzyme-linked immunospot assay using peptides derived from each protein. Our data suggest that a subunit vaccine incorporating bacterially expressed IMV- and EEV-specific proteins can be effective in stimulating anti-vaccinia virus immune responses and providing protection against lethal virus challenge.
Collapse
|
38
|
Abstract
The primary focus of our work is the initiation of an antiviral immune response. While we employ many experimental systems to address this fundamental issue, much of our work revolves around the use of vaccinia virus. Concerns over the negative effects of vaccination have prevented the return of the smallpox immunization program to the general population and underscored the importance of understanding the primary immune response to vaccinia virus. This response is comprised of a complex symphony of immune system components employing a variety of different mechanisms. In this review, we will both highlight the roles of many of these components and touch on the applications of vaccinia virus in the laboratory and the clinic.
Collapse
Affiliation(s)
- Matthew A Fischer
- Department of Microbiology and Immunology, Pennsylvania State University, Milton S. Hershey College of Medicine, Hershey, PA 17033, USA
| | | |
Collapse
|
39
|
Chen Z, Earl P, Americo J, Damon I, Smith SK, Yu F, Sebrell A, Emerson S, Cohen G, Eisenberg RJ, Gorshkova I, Schuck P, Satterfield W, Moss B, Purcell R. Characterization of chimpanzee/human monoclonal antibodies to vaccinia virus A33 glycoprotein and its variola virus homolog in vitro and in a vaccinia virus mouse protection model. J Virol 2007; 81:8989-95. [PMID: 17581986 PMCID: PMC1951440 DOI: 10.1128/jvi.00906-07] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Accepted: 06/11/2007] [Indexed: 01/12/2023] Open
Abstract
Three distinct chimpanzee Fabs against the A33 envelope glycoprotein of vaccinia virus were isolated and converted into complete monoclonal antibodies (MAbs) with human gamma 1 heavy-chain constant regions. The three MAbs (6C, 12C, and 12F) displayed high binding affinities to A33 (K(d) of 0.14 nM to 20 nM) and may recognize the same epitope, which was determined to be conformational and located within amino acid residues 99 to 185 at the C terminus of A33. One or more of the MAbs were shown to reduce the spread of vaccinia virus as well as variola virus (the causative agent of smallpox) in vitro and to more effectively protect mice when administered before or 2 days after intranasal challenge with virulent vaccinia virus than a previously isolated mouse anti-A33 MAb (1G10) or vaccinia virus immunoglobulin. The protective efficacy afforded by anti-A33 MAb was comparable to that of a previously isolated chimpanzee/human anti-B5 MAb. The combination of anti-A33 MAb and anti-B5 MAb did not synergize the protective efficacy. These chimpanzee/human anti-A33 MAbs may be useful in the prevention and treatment of vaccinia virus-induced complications of vaccination against smallpox and may also be effective in the immunoprophylaxis and immunotherapy of smallpox and other orthopoxvirus diseases.
Collapse
Affiliation(s)
- Zhaochun Chen
- Hepatitis Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, 50 South Drive, MSC 8009, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
García AD, Meseda CA, Mayer AE, Kumar A, Merchlinsky M, Weir JP. Characterization and use of mammalian-expressed vaccinia virus extracellular membrane proteins for quantification of the humoral immune response to smallpox vaccines. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2007; 14:1032-44. [PMID: 17596428 PMCID: PMC2044493 DOI: 10.1128/cvi.00050-07] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 05/04/2007] [Accepted: 06/19/2007] [Indexed: 11/20/2022]
Abstract
The licensed smallpox vaccine Dryvax is used as the standard in comparative immunogenicity and protection studies of new smallpox vaccine candidates. Although the correlates of protection against smallpox are unknown, recent studies have shown that a humoral response against the intracellular mature virion and extracellular enveloped virion (EV) forms of vaccinia virus is crucial for protection. Using a recombinant Semliki Forest virus (rSFV) vector system, we expressed a set of full-length EV proteins for the development of EV antigen-specific enzyme-linked immunosorbent assays (ELISAs) and the production of monospecific antisera. The EV-specific ELISAs were used to evaluate the EV humoral response elicited by Dryvax and the nonreplicating modified vaccinia virus Ankara (MVA) in mouse vaccination experiments comparing doses and routes of vaccination. Quantitatively similar titers of antibodies against EV antigens A33R, A56R, and B5R were measured in mice vaccinated with Dryvax and MVA when MVA was administered at a dose of 10(8) plaque-forming units. Further, a substantial increase in the EV-specific antibody response was induced in mice inoculated with MVA by using a prime-boost schedule. Finally, we investigated the abilities of the EV-expressing rSFV vectors to elicit the production of polyclonal monospecific antisera against the corresponding EV proteins in mice. The monospecific serum antibody levels against A33R, A56R, and B5R were measurably higher than the antibody levels induced by Dryvax. The resulting polyclonal antisera were used in Western blot analysis and immunofluorescence assays, indicating that rSFV particles are useful vectors for generating monospecific antisera.
Collapse
Affiliation(s)
- Alonzo D García
- Laboratory of DNA Viruses, Division of Viral Products, Center for Biologics and Evaluation and Research/FDA, 1401 Rockville Pike, HFM-457, Rockville, MD 20892, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Lawrence SJ, Lottenbach KR, Newman FK, Buller RML, Bellone CJ, Chen JJ, Cohen GH, Eisenberg RJ, Belshe RB, Stanley SL, Frey SE. Antibody responses to vaccinia membrane proteins after smallpox vaccination. J Infect Dis 2007; 196:220-9. [PMID: 17570109 PMCID: PMC2533043 DOI: 10.1086/518793] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Accepted: 02/02/2007] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Vaccinia virus (VV) membrane proteins are candidates for orthopoxvirus subunit vaccines and potential targets for therapeutic antibodies. Human antibody responses to these proteins after VV vaccination have not been well characterized. METHODS Pre- and postvaccination (day 26-30) serum specimens from 80 VV vaccine recipients were examined for immunoglobulin G antibodies specific for B5, A33, A27, and L1 by enzyme-linked immunosorbent assay (ELISA). Responses were compared between vaccinia-naive and previously vaccinated (nonnaive) recipients and between nonnaive recipients of undiluted or 1 : 10 diluted vaccine. RESULTS VV vaccination elicited anti-A33 and anti-A27 antibodies in nearly all vaccinia-naive subjects (100% and 93%, respectively). Preexisting antibodies were commonly detected in nonnaive subjects (for anti-B5, 68%; for anti-A33, 59%; for anti-A27, 38%; and for anti-L1, 10%). Anti-B5 antibodies were strongly boosted by undiluted vaccine (geometric mean titer [GMT], 151 vs. 1010 for pre- vs. postvaccination; P<.001), whereas anti-L1 antibody responses were less robust (detection rate, 31%; GMT, 75) in nonnaive subjects. Diluted vaccine elicited antibody responses that were similar to those elicited by undiluted vaccine. CONCLUSIONS Vaccination with VV elicits long-lived specific antibody responses directed against VV membrane proteins that vary by previous vaccination status but not with respect to 10-fold dilution of vaccine. B5, A33, and A27 should be considered for inclusion in future human orthopoxvirus subunit vaccines.
Collapse
Affiliation(s)
- Steven J. Lawrence
- Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri
| | - Kathleen R. Lottenbach
- Division of Infectious Diseases and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri
| | - Frances K. Newman
- Division of Infectious Diseases and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri
| | - R. Mark L. Buller
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri
| | - Clifford J. Bellone
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri
| | - John J. Chen
- Corresponding author for reprints: Steven J. Lawrence, MD Division of Infectious Diseases Washington University School of Medicine Box 8051, 660 South Euclid Avenue St. Louis, Missouri 63110 314−454−8225 (phone) 314−362−9230 (fax)
| | - Gary H. Cohen
- Department of Microbiology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Roselyn J. Eisenberg
- Department of Microbiology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert B. Belshe
- Division of Infectious Diseases and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri
| | - Samuel L. Stanley
- Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri
| | - Sharon E. Frey
- Division of Infectious Diseases and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
42
|
Kim SH, Yeo SG, Park KH, Bang JW, Kim HB, Kim NJ, Jee Y, Cho H, Oh MD, Choe KW. The persistence of humoral and cellular immunities more than three decades after smallpox vaccination. Clin Microbiol Infect 2007; 13:91-3. [PMID: 17184294 DOI: 10.1111/j.1469-0691.2006.01576.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This study assessed the persistence of humoral (neutralising antibody titre to vaccinia virus) and cellular (immediate vaccinia-specific interferon (IFN)-gamma-producing T-cell) immunities to smallpox in a Korean population. Individuals who were vaccinated 25-60 years previously had higher neutralising antibody titres (geometric mean titre (GMT) 13.7; 95% CI 11.0-17.2) than vaccinia-naive individuals (GMT 6.7; 95% CI 5.5-8.0; p <0.001). However, there was no significant difference in cellular immunity between individuals vaccinated previously and vaccinia-naive individuals, and only 15% of the individuals vaccinated previously displayed an immediate IFN-gamma-producing effector-memory response in ELISPOT assays.
Collapse
Affiliation(s)
- S-H Kim
- Center for Infectious Diseases, National Institute of Health, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hayasaka D, Ennis FA, Terajima M. Pathogeneses of respiratory infections with virulent and attenuated vaccinia viruses. Virol J 2007; 4:22. [PMID: 17326843 PMCID: PMC1810241 DOI: 10.1186/1743-422x-4-22] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2006] [Accepted: 02/27/2007] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Respiratory infection with the neurovirulent vaccinia virus (VV) strain Western Reserve (WR) results in an acute infection of the lung followed by dissemination of the virus to other organs and causes lethality in mice. The mechanisms of lethality are not well-understood. In this study, we analyzed virus replication and host immune responses after intranasal infection with lethal and non-lethal doses of VV using the WR strain and the less virulent Wyeth strain. RESULTS The WR strain replicated more vigorously in the lung and in the brain than the Wyeth strain. There were, however, no differences between the virus titers in the brains of mice infected with the higher lethal dose and the lower non-lethal dose of WR strain, suggesting that the amount of virus replication in the brain is unlikely to be the sole determining factor of lethality. The WR strain grew better in primary mouse lung cells than the Wyeth strain. Lethal infection with WR strain was associated with a reduced number of lymphocytes and an altered phenotype of the T cells in the lung compared to non-lethal infections with the WR or Wyeth strains. Severe thymus atrophy with a reduction of CD4 and CD8 double positive T cells was also observed in the lethal infection. CONCLUSION These results suggest that the lethality induced by intranasal infection with a high dose of the WR strain is caused by the higher replication of virus in lung cells and immune suppression during the early phase of the infection, resulting in uncontrolled virus replication in the lung.
Collapse
Affiliation(s)
- Daisuke Hayasaka
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Francis A Ennis
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Masanori Terajima
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
44
|
Fogg CN, Americo JL, Lustig S, Huggins JW, Smith SK, Damon I, Resch W, Earl PL, Klinman DM, Moss B. Adjuvant-enhanced antibody responses to recombinant proteins correlates with protection of mice and monkeys to orthopoxvirus challenges. Vaccine 2007; 25:2787-99. [PMID: 17229505 PMCID: PMC1952660 DOI: 10.1016/j.vaccine.2006.12.037] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2006] [Revised: 11/27/2006] [Accepted: 12/13/2006] [Indexed: 12/04/2022]
Abstract
Recombinant proteins are being evaluated as smallpox and monkeypox vaccines because of their perceived safety compared to live vaccinia virus. Previously, we demonstrated that three or more injections of a Ribi-type adjuvant with a combination of three proteins from the outer membranes of intracellular (L1 protein) and extracellular (A33 and B5 proteins) forms of vaccinia virus protected mice against a lethal intranasal challenge with vaccinia virus. Here, we compared several adjuvants and found that QS-21 and to a lesser extent alum + CpG oligodeoxynucleotides accelerated and enhanced neutralizing antibody responses to a mixture of L1 and A33 proteins, provided the highest ratio of IgG2a to IgG1 isotype response, and protected mice against disease and death after only two immunizations 3 weeks apart. In addition, monkeys immunized with recombinant vaccinia virus proteins and QS-21 developed neutralizing antibody to monkeypox virus and had reduced virus load, skin lesions, and morbidity compared to the non-immunized group following monkeypox virus challenge.
Collapse
Affiliation(s)
- Christiana N Fogg
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The smallpox vaccine consists of live vaccinia virus and is generally considered the gold standard of vaccines, since it is the only one that has led to the complete eradication of an infectious disease from the human population. Renewed fears that smallpox might be deliberately released in an act of bioterrorism have led to resurgence in the study of immunity and immunological memory to vaccinia virus and other poxviruses. Here we review our current understanding of memory T-cell, memory B-cell, and antibody responses to vaccinia and related poxviruses, both in animal models and human subjects. Of particular interest are recent advances in understanding protective immunity to poxviruses, quantifying immunological memory to the smallpox vaccine in humans, and identifying major vaccinia-specific T-cell and B-cell epitopes. In addition, potential mechanisms for maintenance of immunological memory are discussed.
Collapse
Affiliation(s)
- Ian J Amanna
- OHSU Vaccine and Gene Therapy Institute, Beaverton, OR, USA
| | | | | |
Collapse
|
46
|
Tang J, Murtadha M, Schnell M, Eisenlohr LC, Hooper J, Flomenberg P. Human T-cell responses to vaccinia virus envelope proteins. J Virol 2006; 80:10010-20. [PMID: 17005679 PMCID: PMC1617304 DOI: 10.1128/jvi.00601-06] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
One approach for a safer smallpox vaccine is to utilize recombinant subunits rather than live vaccinia virus (VACV). The products of the VACV envelope genes A27L, L1R, B5R, and A33R induce protective antibodies in animal models. We propose that proteins that elicit T-cell responses, as well as neutralizing antibodies, will be important to include in a molecular vaccine. To evaluate VACV-specific memory T-cell responses, peripheral blood mononuclear cells (PBMC) from four VACV vaccinees were tested against whole VACV and the individual envelope proteins A27, B5, L1, and A33, using gamma interferon enzyme-linked immunospot and cytokine flow cytometry assays. PBMC were stimulated with autologous dendritic cells infected with VACV or electroporated with individual VACV protein mRNAs. T-cell lines from all donors, vaccinated from 1 month to over 20 years ago, recognized all four VACV envelope proteins. Both CD4(+) and CD8(+) T-cell responses to each protein were detected. Further analysis focused on representative proteins B5 and A27. PBMC from a recent vaccinee exhibited high frequencies of CD4(+) and CD8(+) T-cell precursors to both B5 (19.8 and 20%, respectively) and A27 (6.8 and 3.7%). In comparison, B5- and A27-specific T-cell frequencies ranged from 0.4 to 1.3% in a donor vaccinated 3 years ago. Multiple CD4(+) and CD8(+) T-cell epitopes were identified from both A27 and B5, using overlapping 15-mer peptides. These data suggest that all four VACV envelope proteins may contribute to protective immunity, not only by inducing antibody responses, but also by eliciting T-cell responses.
Collapse
Affiliation(s)
- Jie Tang
- Thomas Jefferson University, 1020 Locust Street, Rm. 329, Philadelphia, PA 19107, USA
| | | | | | | | | | | |
Collapse
|
47
|
Pütz MM, Midgley CM, Law M, Smith GL. Quantification of antibody responses against multiple antigens of the two infectious forms of Vaccinia virus provides a benchmark for smallpox vaccination. Nat Med 2006; 12:1310-5. [PMID: 17086190 DOI: 10.1038/nm1457] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2005] [Accepted: 06/23/2006] [Indexed: 01/29/2023]
Abstract
Smallpox was eradicated without an adequate understanding of how vaccination induced protection. In response to possible bioterrorism with smallpox, the UK government vaccinated approximately 300 health care workers with vaccinia virus (VACV) strain Lister. Antibody responses were analyzed using ELISA for multiple surface antigens of the extracellular enveloped virus (EEV) and the intracellular mature virus (IMV), plaque reduction neutralization and a fluorescence-based flow cytometric neutralization assay. Antibody depletion experiments showed that the EEV surface protein B5 is the only target responsible for EEV neutralization in vaccinated humans, whereas multiple IMV surface proteins, including A27 and H3, are targets for IMV-neutralizing antibodies. These data suggest that it would be unwise to exclude the B5 protein from a future smallpox vaccine. Repeated vaccination provided significantly higher B5-specific and thus EEV-neutralizing antibody responses. These data provide a benchmark against which new, safer smallpox vaccines and residual immunity can be compared.
Collapse
Affiliation(s)
- Mike M Pütz
- Department of Virology, Faculty of Medicine, Imperial College London, St. Mary's Campus, London W2 1PG, UK
| | | | | | | |
Collapse
|
48
|
Staib C, Suezer Y, Kisling S, Kalinke U, Sutter G. Short-term, but not post-exposure, protection against lethal orthopoxvirus challenge after immunization with modified vaccinia virus Ankara. J Gen Virol 2006; 87:2917-2921. [PMID: 16963750 DOI: 10.1099/vir.0.82068-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Safety-tested vaccinia virus (VACV) MVA serves as a candidate third-generation vaccine against smallpox. Here, MVA immunization of mice shortly before or after lethal respiratory challenge with VACV Western Reserve was investigated. Whilst post-exposure treatment failed to protect animals, immunizations on day 2 prior to challenge were fully protective. On the day of challenge, MVA inoculation may prevent death, but not onset of severe respiratory disease. After intranasal MVA application, massive influx of leukocytes (such as neutrophils, macrophages, natural killer cells and T cells) was found in the lungs of the animals, indicating the contribution of innate responses to protection. Correspondingly, in RAG-1-/- mice, MVA inoculation delayed onset of disease significantly, but did not prevent fatal infection. Thus, short-term protection required a tight interplay of both innate and adaptive antiviral immunity. These data suggest that, in addition to conventional vaccination, MVA may serve for potent emergency prophylaxis against orthopoxvirus infection.
Collapse
Affiliation(s)
- Caroline Staib
- GSF - Institute of Molecular Virology and Institute of Virology, Technical University of Munich, 81675 München, Germany
| | - Yasemin Suezer
- Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany
| | - Sigrid Kisling
- GSF - Institute of Molecular Virology and Institute of Virology, Technical University of Munich, 81675 München, Germany
| | - Ulrich Kalinke
- Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany
| | - Gerd Sutter
- Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany
| |
Collapse
|
49
|
Law M, Carter GC, Roberts KL, Hollinshead M, Smith GL. Ligand-induced and nonfusogenic dissolution of a viral membrane. Proc Natl Acad Sci U S A 2006; 103:5989-94. [PMID: 16585508 PMCID: PMC1424662 DOI: 10.1073/pnas.0601025103] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Indexed: 11/18/2022] Open
Abstract
Hitherto, all enveloped viruses were thought to shed their lipid membrane during entry into cells by membrane fusion. The extracellular form of Vaccinia virus has two lipid envelopes surrounding the virus core, and consequently a single fusion event will not deliver a naked core into the cell. Here we report a previously underscribed mechanism in which the outer viral membrane is disrupted by a ligand-induced nonfusogenic reaction, followed by the fusion of the inner viral membrane with the plasma membrane and penetration of the virus core into the cytoplasm. The dissolution of the outer envelope depends on interactions with cellular polyanionic molecules and requires the virus glycoproteins A34 and B5. This discovery represents a remarkable example of how viruses manipulate biological membranes, solves the topological problem of how a double-enveloped virus enters cells, reveals a new effect of polyanions on viruses, and provides a therapeutic approach for treatment of poxvirus infections, such as smallpox.
Collapse
Affiliation(s)
- Mansun Law
- Department of Virology, Faculty of Medicine, Imperial College London, St. Mary’s Campus, Norfolk Place, London W2 1PG, United Kingdom
| | - Gemma C. Carter
- Department of Virology, Faculty of Medicine, Imperial College London, St. Mary’s Campus, Norfolk Place, London W2 1PG, United Kingdom
| | - Kim L. Roberts
- Department of Virology, Faculty of Medicine, Imperial College London, St. Mary’s Campus, Norfolk Place, London W2 1PG, United Kingdom
| | - Michael Hollinshead
- Department of Virology, Faculty of Medicine, Imperial College London, St. Mary’s Campus, Norfolk Place, London W2 1PG, United Kingdom
| | - Geoffrey L. Smith
- Department of Virology, Faculty of Medicine, Imperial College London, St. Mary’s Campus, Norfolk Place, London W2 1PG, United Kingdom
| |
Collapse
|
50
|
Chen Z, Earl P, Americo J, Damon I, Smith SK, Zhou YH, Yu F, Sebrell A, Emerson S, Cohen G, Eisenberg RJ, Svitel J, Schuck P, Satterfield W, Moss B, Purcell R. Chimpanzee/human mAbs to vaccinia virus B5 protein neutralize vaccinia and smallpox viruses and protect mice against vaccinia virus. Proc Natl Acad Sci U S A 2006; 103:1882-7. [PMID: 16436502 PMCID: PMC1413659 DOI: 10.1073/pnas.0510598103] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Chimpanzee Fabs against the B5 envelope glycoprotein of vaccinia virus were isolated and converted into complete mAbs with human gamma 1 heavy chain constant regions. The two mAbs (8AH8AL and 8AH7AL) displayed high binding affinities to B5 (Kd of 0.2 and 0.7 nM). The mAb 8AH8AL inhibited the spread of vaccinia virus as well as variola virus (the causative agent of smallpox) in vitro, protected mice from subsequent intranasal challenge with virulent vaccinia virus, protected mice when administered 2 days after challenge, and provided significantly greater protection than that afforded by a previously isolated rat anti-B5 mAb (19C2) or by vaccinia immune globulin. The mAb bound to a conformational epitope between amino acids 20 and 130 of B5. These chimpanzee/human anti-B5 mAbs may be useful in the prevention and treatment of vaccinia virus-induced complications of vaccination against smallpox and may also be effective in the immunoprophylaxis and immunotherapy of smallpox.
Collapse
Affiliation(s)
| | - Patricia Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, and
| | - Jeffrey Americo
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, and
| | - Inger Damon
- Centers for Disease Control and Prevention, Atlanta, GA 30333
| | - Scott K. Smith
- Centers for Disease Control and Prevention, Atlanta, GA 30333
| | | | | | | | - Suzanne Emerson
- Molecular Hepatitis Section, Laboratory of Infectious Diseases, and
| | - Gary Cohen
- Department of Microbiology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Roselyn J. Eisenberg
- Department of Microbiology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Juraj Svitel
- **Protein Biophysics Resource, Office of Research Services, Office of the Director, National Institutes of Health, Bethesda, MD 20892
| | - Peter Schuck
- **Protein Biophysics Resource, Office of Research Services, Office of the Director, National Institutes of Health, Bethesda, MD 20892
| | - William Satterfield
- Department of Veterinary Sciences, University of Texas M. D. Anderson Cancer Center, Bastrop, TX 78602
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, and
| | - Robert Purcell
- *Hepatitis Viruses Section
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|