1
|
Xu Q, Zhang RR, Wu M, Zhang J, Wang ZX, Chi H, Zhou C, Xiong XC, Liu HT, Qin CF, Ye Q. A Single-Chain Mpox mRNA Vaccine Elicits Protective Immune Response in Mice. Vaccines (Basel) 2025; 13:514. [PMID: 40432124 PMCID: PMC12115521 DOI: 10.3390/vaccines13050514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2025] [Revised: 04/28/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Background: The re-emerging mpox virus (MPXV) has spread to numerous countries and raised global concern. There is an urgent need for a safe and effective mRNA vaccine candidate against MPXV infection. Previously, we developed a penta-component mRNA vaccine that contained five distinct antigen-encoded mRNAs encapsulated within lipid nanoparticles (LNPs). Here, we sought to develop a single-chain mRNA vaccine that encodes antigens derived from both intracellular mature virion (IMV) and extracellular enveloped virion (EEV). Methods: A single-chain mRNA vaccine encoding a fusion protein comprising the ectodomains of M1R (eM1R) and A35R (eA35R) (MPXVeM1-eA35) was developed and characterized, while an admixed formulation of two individual mRNA-LNPs encoding separate antigens was developed as the control (MPXVeM1+eA35). Meanwhile, based on the same strategy, we designed a single-chain mRNA vaccine encoding dimeric antigens (MPXVeM1-eA35-Fc). Mice were immunized with two doses of the candidate vaccines, and both humoral and cellular immune responses were evaluated. The protective efficacy of the candidate vaccines was evaluated based on body weight monitoring and tissue viral load measurement after challenge with vaccinia virus (VACV). Results: Immunization with two doses of MPXVeM1-eA35 elicited robust levels of neutralizing antibodies and antigen-specific cellular immune response. Importantly, MPXVeM1-eA35 demonstrated protective efficacy in a VACV challenge mouse model and showed superior capacity in preventing weight loss post-challenge compared to MPXVeM1+eA35. Similarly, MPXVeM1-eA35-Fc exhibited comparable or superior immunogenicity and protective efficacy compared to the admixed formulations. Conclusions: The single-chain mRNA vaccine elicited a protective immune response in mice, offering significant advantages in terms of manufacturing processes and quality control. Our single-chain mRNA vaccine platform presents a promising strategy for the next generation design of mpox vaccines and contributes to the mitigation of MPXV endemic worldwide.
Collapse
Affiliation(s)
- Qian Xu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Q.X.); (J.Z.)
| | - Rong-Rong Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (R.-R.Z.); (M.W.); (Z.-X.W.); (H.C.); (C.Z.); (X.-C.X.); (H.-T.L.)
| | - Mei Wu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (R.-R.Z.); (M.W.); (Z.-X.W.); (H.C.); (C.Z.); (X.-C.X.); (H.-T.L.)
| | - Jie Zhang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Q.X.); (J.Z.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (R.-R.Z.); (M.W.); (Z.-X.W.); (H.C.); (C.Z.); (X.-C.X.); (H.-T.L.)
| | - Zu-Xin Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (R.-R.Z.); (M.W.); (Z.-X.W.); (H.C.); (C.Z.); (X.-C.X.); (H.-T.L.)
| | - Hang Chi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (R.-R.Z.); (M.W.); (Z.-X.W.); (H.C.); (C.Z.); (X.-C.X.); (H.-T.L.)
| | - Chao Zhou
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (R.-R.Z.); (M.W.); (Z.-X.W.); (H.C.); (C.Z.); (X.-C.X.); (H.-T.L.)
| | - Xiao-Chuan Xiong
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (R.-R.Z.); (M.W.); (Z.-X.W.); (H.C.); (C.Z.); (X.-C.X.); (H.-T.L.)
| | - Hai-Tao Liu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (R.-R.Z.); (M.W.); (Z.-X.W.); (H.C.); (C.Z.); (X.-C.X.); (H.-T.L.)
| | - Cheng-Feng Qin
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Q.X.); (J.Z.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (R.-R.Z.); (M.W.); (Z.-X.W.); (H.C.); (C.Z.); (X.-C.X.); (H.-T.L.)
| | - Qing Ye
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (R.-R.Z.); (M.W.); (Z.-X.W.); (H.C.); (C.Z.); (X.-C.X.); (H.-T.L.)
| |
Collapse
|
2
|
Link EK, Tscherne A, Sutter G, Smith ER, Gurwith M, Chen RT, Volz A. A Brighton collaboration standardized template with key considerations for a benefit/risk assessment for a viral vector vaccine based on a non-replicating modified vaccinia virus Ankara viral vector. Vaccine 2025; 43:126521. [PMID: 39612556 DOI: 10.1016/j.vaccine.2024.126521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
The Brighton Collaboration Benefit-Risk Assessment of VAccines by TechnolOgy (BRAVATO) was formed to evaluate the safety and other key features of new platform technology vaccines. This manuscript provides an overview of Modified Vaccinia virus Ankara (MVA)-vectored vaccines and reviews molecular and biological key features of this platform. In particular, this review aims to provide fundamental information about the promising candidate vaccine MVA-MERS-S which has been evaluated successfully in different preclinical animal models and has undergone clinical testing including a phase Ib study involving more than 170 participants. Infectious diseases continue to be a major cause of human death worldwide. In this context, emerging zoonotic infectious diseases pose a particular challenge for public health systems. In the last two decades, three different respiratory coronaviruses, including the Middle East respiratory syndrome Coronavirus (MERS-CoV) have emerged. For many years, safe and efficacious vaccines have been a major tool to combat infectious diseases. Here, we report on a promising candidate vaccine (MVA-MERS-S) against MERS-CoV based on MVA. Upon application, MVA-MERS-S has been well tolerated and immunogenic, inducing both, cellular and humoral immune responses in different animal models and humans. We demonstrate that the MVA vector platform, with the example of MVA-MERS-S, is a viable and effective tool for producing safe, immunogenic, and efficient vaccines against emerging infectious diseases.
Collapse
Affiliation(s)
- Ellen K Link
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Alina Tscherne
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Gerd Sutter
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Emily R Smith
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA.
| | - Marc Gurwith
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| | - Robert T Chen
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| | - Asisa Volz
- University of Veterinary Medicine Hannover, Institute of Virology, Hannover, Germany
| |
Collapse
|
3
|
Wen Y, Deng S, Wang T, Gao M, Nan W, Tang F, Xue Q, Ju Y, Dai J, Wei Y, Xue F. Novel strategy for Poxviridae prevention: Thermostable combined subunit vaccine patch with intense immune response. Antiviral Res 2024; 228:105943. [PMID: 38909959 DOI: 10.1016/j.antiviral.2024.105943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Poxviruses gained international attention due to the sharp rise in monkeypox cases in recent years, highlighting the urgent need for the development of a secure and reliable vaccine. This study involved the development of an innovative combined subunit vaccine (CSV) targeting poxviruses, with lumpy skin disease virus (LSDV) serving as the model virus. To this end, the potential sites for poxvirus vaccines were fully evaluated to develop and purify four recombinant proteins. These proteins were then successfully delivered to the dermis in a mouse model by utilizing dissolvable microneedle patches (DMPs). This approach simplified the vaccination procedure and significantly mitigated the associated risk. CSV-loaded DMPs contained four recombinant proteins and a novel adjuvant, CpG, which allowed DMPs to elicit the same intensity of humoral and cellular immunity as subcutaneous injection. Following immunization with SC and DMP, the mice exhibited notable levels of neutralizing antibodies, albeit at a low concentration. It is noteworthy that the CSV loaded into DMPs remained stable for at least 4 months at room temperature, effectively addressing the storage and transportation challenges. Based on the study findings, CSV-loaded DMPs are expected to be utilized worldwide as an innovative technique for poxvirus inoculation, especially in underdeveloped regions. This novel strategy is crucial for the development of future poxvirus vaccines.
Collapse
MESH Headings
- Animals
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Mice
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Poxviridae Infections/prevention & control
- Poxviridae Infections/immunology
- Female
- Poxviridae/immunology
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- Mice, Inbred BALB C
- Lumpy skin disease virus/immunology
- Vaccination
- Immunity, Cellular
- Immunity, Humoral
- Recombinant Proteins/immunology
- Recombinant Proteins/administration & dosage
- Adjuvants, Vaccine/administration & dosage
- Adjuvants, Immunologic/administration & dosage
Collapse
Affiliation(s)
- Yuan Wen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Sanya Institute of Nanjing Agricultural University, Sanya, 572025, China
| | - Shuyue Deng
- College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Tianmin Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Sanya Institute of Nanjing Agricultural University, Sanya, 572025, China
| | - Mengtian Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Sanya Institute of Nanjing Agricultural University, Sanya, 572025, China
| | - Wenlong Nan
- Laboratory of Diagnostics Development, China Animal Health and Epidemiology Center, 369 Nanjing Road, Qingdao, 266032, China
| | - Fang Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing, 100081, China
| | - Yanmin Ju
- College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jianjun Dai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yurong Wei
- Xinjiang Key Laboratory of Animal Infectious Diseases, Institute of Veterinary Medicine, Xinjiang Academy of Animal Science, Urumqi, 830099, China
| | - Feng Xue
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Sanya Institute of Nanjing Agricultural University, Sanya, 572025, China.
| |
Collapse
|
4
|
Branda F, Romano C, Ciccozzi M, Giovanetti M, Scarpa F, Ciccozzi A, Maruotti A. Mpox: An Overview of Pathogenesis, Diagnosis, and Public Health Implications. J Clin Med 2024; 13:2234. [PMID: 38673507 PMCID: PMC11050819 DOI: 10.3390/jcm13082234] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Mpox, caused by viruses of the genus Orthopoxvirus, is an emerging threat to human and animal health. With increasing urbanization and more frequent interaction between humans and wild animals, the risk of Mpox transmission to humans has increased significantly. This review aims to examine in depth the epidemiology, pathogenesis, and diagnosis of Mpox, with a special focus on recent discoveries and advances in understanding the disease. Molecular mechanisms involved in viral replication will be examined, as well as risk factors associated with interspecific transmission and spread of the disease in human populations. Currently available diagnostic methods will also be discussed, with a critical analysis of their limitations and possible future directions for improving the accuracy and timeliness of diagnosis. Finally, this review will explore the public health implications associated with Mpox, emphasizing the importance of epidemiological surveillance, vaccination, and emergency preparedness to prevent and manage possible outbreaks. Understanding the epidemiology and control strategies for Mpox is critical to protecting the health of human and animal communities and mitigating the risk of interspecific transmission and spread of the disease.
Collapse
Affiliation(s)
- Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (F.B.)
| | - Chiara Romano
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (F.B.)
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (F.B.)
| | - Marta Giovanetti
- Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
- Climate Amplified Diseases and Epidemics (CLIMADE), Brasilia 70070-130, Brazil
- Instituto Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil
| | - Fabio Scarpa
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43b, 07100 Sassari, Italy
| | - Alessandra Ciccozzi
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43b, 07100 Sassari, Italy
| | | |
Collapse
|
5
|
Sang Y, Zhang Z, Liu F, Lu H, Yu C, Sun H, Long J, Cao Y, Mai J, Miao Y, Wang X, Fang J, Wang Y, Huang W, Yang J, Wang S. Monkeypox virus quadrivalent mRNA vaccine induces immune response and protects against vaccinia virus. Signal Transduct Target Ther 2023; 8:172. [PMID: 37117161 PMCID: PMC10144886 DOI: 10.1038/s41392-023-01432-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/19/2023] [Accepted: 03/31/2023] [Indexed: 04/30/2023] Open
Abstract
Monkeypox has been declared a public health emergency by the World Health Organization. There is an urgent need for efficient and safe vaccines against the monkeypox virus (MPXV) in response to the rapidly spreading monkeypox epidemic. In the age of COVID-19, mRNA vaccines have been highly successful and emerged as platforms enabling rapid development and large-scale preparation. Here, we develop two MPXV quadrivalent mRNA vaccines, named mRNA-A-LNP and mRNA-B-LNP, based on two intracellular mature virus specific proteins (A29L and M1R) and two extracellular enveloped virus specific proteins (A35R and B6R). By administering mRNA-A-LNP and mRNA-B-LNP intramuscularly twice, mice induce MPXV specific IgG antibodies and potent vaccinia virus (VACV) specific neutralizing antibodies. Further, it elicits efficient MPXV specific Th-1 biased cellular immunity, as well as durable effector memory T and germinal center B cell responses in mice. In addition, two doses of mRNA-A-LNP and mRNA-B-LNP are protective against the VACV challenge in mice. And, the passive transfer of sera from mRNA-A-LNP and mRNA-B-LNP-immunized mice protects nude mice against the VACV challenge. Overall, our results demonstrate that mRNA-A-LNP and mRNA-B-LNP appear to be safe and effective vaccine candidates against monkeypox epidemics, as well as against outbreaks caused by other orthopoxviruses, including the smallpox virus.
Collapse
Affiliation(s)
- Ye Sang
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China
| | - Zhen Zhang
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China
| | - Fan Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, P. R. China
| | - Haitao Lu
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China
| | - Changxiao Yu
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China
| | - Huisheng Sun
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China
| | - Jinrong Long
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China
| | - Yiming Cao
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China
| | - Jierui Mai
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China
| | - Yiqi Miao
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China
| | - Xin Wang
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China
| | - Jiaxin Fang
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China
| | - Youchun Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, 650031, P. R. China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, P. R. China.
| | - Jing Yang
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China.
| | - Shengqi Wang
- Bioinformatics center of AMMS, Beijing, 100850, P. R. China.
| |
Collapse
|
6
|
A nucleic acid-based orthopoxvirus vaccine targeting the vaccinia virus L1, A27, B5 and A33 proteins protects rabbits against lethal rabbitpox virus aerosol challenge. J Virol 2021; 96:e0150421. [PMID: 34851148 DOI: 10.1128/jvi.01504-21] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In the age of COVID, nucleic acid vaccines have garnered much attention, at least in part, because of the simplicity of construction, production, and flexibility to adjust and adapt to an evolving outbreak. Orthopoxviruses remain a threat on multiple fronts, especially as emerging zoonosis. In response, we developed a DNA vaccine, termed 4pox, that protected nonhuman primates against monkeypox virus (MPXV) induced severe disease. Here, we examined the protective efficacy of the 4pox DNA vaccine delivered by intramuscular (i.m.) electroporation (EP) in rabbits challenged with aerosolized rabbitpox virus (RPXV), a model that recapitulates the respiratory route of exposure and low dose associated with natural smallpox exposure in humans. We found that 4pox vaccinated rabbits developed immunogen-specific antibodies, including neutralizing antibodies and did not develop any clinical disease, indicating protection against aerosolized RPXV. In contrast, unvaccinated animals developed significant signs of disease, including lesions, and were euthanized. These findings demonstrate that an unformulated, non-adjuvanted DNA vaccine delivered (i.m.) can protect against an aerosol exposure. Importance The eradication of smallpox and subsequent cessation of vaccination has left a majority of the population susceptible to variola virus or other emerging poxvirus. This is exemplified by human monkeypox, as evidenced by the increase in reported endemic and imported cases over the past decades. Therefore, a malleable vaccine technology that can be mass produced, and doesn't require complex conditions for distribution and storage is sought. Herein, we show that a DNA vaccine, in the absence of a specialized formulation or adjuvant, can protect against a lethal aerosol insult of rabbitpox virus.
Collapse
|
7
|
Lei V, Petty AJ, Atwater AR, Wolfe SA, MacLeod AS. Skin Viral Infections: Host Antiviral Innate Immunity and Viral Immune Evasion. Front Immunol 2020; 11:593901. [PMID: 33240281 PMCID: PMC7677409 DOI: 10.3389/fimmu.2020.593901] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/06/2020] [Indexed: 12/16/2022] Open
Abstract
The skin is an active immune organ that functions as the first and largest site of defense to the outside environment. Serving as the primary interface between host and pathogen, the skin’s early immune responses to viral invaders often determine the course and severity of infection. We review the current literature pertaining to the mechanisms of cutaneous viral invasion for classical skin-tropic, oncogenic, and vector-borne skin viruses. We discuss the skin’s evolved mechanisms for innate immune viral defense against these invading pathogens, as well as unique strategies utilized by the viruses to escape immune detection. We additionally explore the roles that demographic and environmental factors, such as age, biological sex, and the cutaneous microbiome, play in altering the host immune response to viral threats.
Collapse
Affiliation(s)
- Vivian Lei
- Department of Dermatology, Duke University, Durham, NC, United States.,School of Medicine, Duke University, Durham, NC, United States
| | - Amy J Petty
- School of Medicine, Duke University, Durham, NC, United States
| | - Amber R Atwater
- Department of Dermatology, Duke University, Durham, NC, United States
| | - Sarah A Wolfe
- Department of Dermatology, Duke University, Durham, NC, United States
| | - Amanda S MacLeod
- Department of Dermatology, Duke University, Durham, NC, United States.,Department of Immunology, Duke University, Durham, NC, United States.,Pinnell Center for Investigative Dermatology, Duke University, Durham, NC, United States.,Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| |
Collapse
|
8
|
Li Y, Chen S, Fang J, Zhu Y, Bai B, Li W, Yin X, Wang J, Liu X, Han J, Li X, Sun L, Jin N. Construction of an attenuated Tian Tan vaccinia virus strain by deletion of TA35R and TJ2R genes. Virus Res 2018; 256:192-200. [PMID: 30190251 DOI: 10.1016/j.virusres.2018.06.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/29/2018] [Accepted: 06/30/2018] [Indexed: 12/19/2022]
Abstract
rVTT-TA35-TJ, an attenuated vaccinia virus Tian Tan strain (VTT), was constructed by knocking out two non-essential gene fragments (TA35R and TJ2R) related to virulence, immunomodulation, and host range; and by combining double marker screening with exogenous and endogenous selectable marker knock-out techniques. Here, the shuttle plasmids pSK-TA35 and pSK-TJ were constructed, containing two pairs of recombinant arms: early and late strong promoter pE/L and EGFP as an exogenous selectable marker. The recombinant vaccinia virus rVTT-TA35-TJ without exogenous selection markers was then obtained through homologous recombination technology and the Cre/loxP system. Knocking out the two gene fragments does not affect the replication ability of the virus and displays a good genetic stability. Furthermore, a series of in vivo and in vitro experiments demonstrate that although virulence of rVTT-TA35-TJ is attenuated significantly, high immunogenicity was maintained. These results support the potential development of rVTT-TA35-TJ as a safe viral vector or vaccine.
Collapse
Affiliation(s)
- Yiquan Li
- Medical College, Yanbian University, Yanji, 133002, PR China; Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China
| | - Shuang Chen
- Medical College, Yanbian University, Yanji, 133002, PR China; Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China; School of Medical Inspection, Jilin Medical University, Jilin, 132013, PR China
| | - Jinbo Fang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China; Changchun University of Chinese Medicine, Changchun, 130021, PR China
| | - Yilong Zhu
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China; Changchun University of Chinese Medicine, Changchun, 130021, PR China
| | - Bing Bai
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China; Changchun University of Chinese Medicine, Changchun, 130021, PR China
| | - Wenjie Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China; Changchun University of Chinese Medicine, Changchun, 130021, PR China; College of Animal Science and Technology, Guangxi University, Nanning, 530005, PR China
| | - Xunzhe Yin
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China; Changchun University of Chinese Medicine, Changchun, 130021, PR China
| | - Jing Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China
| | - Xing Liu
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China
| | - Jicheng Han
- Medical College, Yanbian University, Yanji, 133002, PR China; Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China
| | - Xiao Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China; Changchun University of Chinese Medicine, Changchun, 130021, PR China; Institute of Virology, Wenzhou University Town, Wenzhou, 325035, PR China.
| | - Lili Sun
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China; Department of Head and Neck Surgery, Tumor Hospital of Jilin Province, Changchun, 130012, PR China.
| | - Ningyi Jin
- Medical College, Yanbian University, Yanji, 133002, PR China; Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, 130122, PR China; Changchun University of Chinese Medicine, Changchun, 130021, PR China; Institute of Virology, Wenzhou University Town, Wenzhou, 325035, PR China; Jiang su Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China.
| |
Collapse
|
9
|
Development of an animal model of progressive vaccinia in nu/nu mice and the use of bioluminescence imaging for assessment of the efficacy of monoclonal antibodies against vaccinial B5 and L1 proteins. Antiviral Res 2017; 144:8-20. [PMID: 28495463 DOI: 10.1016/j.antiviral.2017.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 11/24/2022]
Abstract
Bioluminescence imaging (BLI) was used to follow dissemination of recombinant vaccinia virus (VACV) expressing luciferase (IHD-J-Luc) in BALB/c nu/nu mice treated post-challenge with monoclonal antibodies (MAbs) against L1 and B5 VACV proteins in a model of Progressive Vaccinia (PV). Areas Under the flux Curve (AUC) were calculated for viral loads in multiple organs in individual mice. Following scarification with 105 pfu, IHD-J-Luc VACV undergoes fast replication at the injection site and disseminates rapidly to the inguinal lymph nodes followed by spleen, liver, and axillary lymph nodes within 2-3 days and before primary lesions are visible at the site of scarification. Extension of survival in nude mice treated with a combination of anti-B5 and anti-L1 MAbs 24 h post challenge correlated with a significant reduction in viral load at the site of scarification and delayed systemic dissemination. Nude mice reconstituted with 104 T cells prior to challenge with IHD-J-Luc, and treated with MAbs post-challenge, survived infection, cleared the virus from all organs and scarification site, and developed anti-VACV IgG and VACV-specific polyfunctional CD8+ T cells that co-expressed the degranulation marker CD107a, and IFNγ and TNFα cytokines. All T cell reconstituted mice survived intranasal re-challenge with IHD-J-Luc (104 pfu) two months after the primary infection. Thus, using BLI to monitor VACV replication in a PV model, we showed that anti-VACV MAbs administered post challenge extended survival of nude mice and protected T cell reconstituted nude mice from lethality by reducing replication at the site of scarification and systemic dissemination of VACV.
Collapse
|
10
|
Bissa M, Quaglino E, Zanotto C, Illiano E, Rolih V, Pacchioni S, Cavallo F, De Giuli Morghen C, Radaelli A. Protection of mice against the highly pathogenic VV IHD-J by DNA and fowlpox recombinant vaccines, administered by electroporation and intranasal routes, correlates with serum neutralizing activity. Antiviral Res 2016; 134:182-191. [PMID: 27637905 PMCID: PMC9533953 DOI: 10.1016/j.antiviral.2016.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/05/2016] [Accepted: 09/09/2016] [Indexed: 11/06/2022]
Abstract
The control of smallpox was achieved using live vaccinia virus (VV) vaccine, which successfully eradicated the disease worldwide. As the variola virus no longer exists as a natural infection agent, mass vaccination was discontinued after 1980. However, emergence of smallpox outbreaks caused by accidental or deliberate release of variola virus has stimulated new research for second-generation vaccine development based on attenuated VV strains. Considering the closely related animal poxviruses that also arise as zoonoses, and the increasing number of unvaccinated or immunocompromised people, a safer and more effective vaccine is still required. With this aim, new vectors based on avian poxviruses that cannot replicate in mammals should improve the safety of conventional vaccines, and protect from zoonotic orthopoxvirus diseases, such as cowpox and monkeypox. In this study, DNA and fowlpox (FP) recombinants that expressed the VV L1R, A27L, A33R, and B5R genes were generated (4DNAmix, 4FPmix, respectively) and tested in mice using novel administration routes. Mice were primed with 4DNAmix by electroporation, and boosted with 4FPmix applied intranasally. The lethal VVIHD-J strain was then administered by intranasal challenge. All of the mice receiving 4DNAmix followed by 4FPmix, and 20% of the mice immunized only with 4FPmix, were protected. The induction of specific humoral and cellular immune responses directly correlated with this protection. In particular, higher anti-A27 antibodies and IFNγ-producing T lymphocytes were measured in the blood and spleen of the protected mice, as compared to controls. VVIHD-J neutralizing antibodies in sera from the protected mice suggest that the prime/boost vaccination regimen with 4DNAmix plus 4FPmix may be an effective and safe mode to induce protection against smallpox and poxvirus zoonotic infections. The electroporation/intranasal administration routes contributed to effective immune responses and mouse survival.
Collapse
Affiliation(s)
- Massimiliano Bissa
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, 20133 Milano, Italy.
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Torino, Italy.
| | - Carlo Zanotto
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Via Vanvitelli, 32, 20129 Milano, Italy.
| | - Elena Illiano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, 20133 Milano, Italy.
| | - Valeria Rolih
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Torino, Italy.
| | - Sole Pacchioni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, 20133 Milano, Italy.
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza 52, 10126 Torino, Italy.
| | - Carlo De Giuli Morghen
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Via Vanvitelli, 32, 20129 Milano, Italy; Catholic University "Our Lady of Good Counsel", Rr. Dritan Hoxha, Tirana, Albania.
| | - Antonia Radaelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, 20133 Milano, Italy; Cellular and Molecular Pharmacology Section, National Research Council (CNR), Institute of Neurosciences, University of Milan, Via Vanvitelli, 32, 20129 Milano, Italy.
| |
Collapse
|
11
|
Stern D, Pauly D, Zydek M, Miller L, Piesker J, Laue M, Lisdat F, Dorner MB, Dorner BG, Nitsche A. Development of a Genus-Specific Antigen Capture ELISA for Orthopoxviruses - Target Selection and Optimized Screening. PLoS One 2016; 11:e0150110. [PMID: 26930499 PMCID: PMC4773239 DOI: 10.1371/journal.pone.0150110] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/09/2016] [Indexed: 11/18/2022] Open
Abstract
Orthopoxvirus species like cowpox, vaccinia and monkeypox virus cause zoonotic infections in humans worldwide. Infections often occur in rural areas lacking proper diagnostic infrastructure as exemplified by monkeypox, which is endemic in Western and Central Africa. While PCR detection requires demanding equipment and is restricted to genome detection, the evidence of virus particles can complement or replace PCR. Therefore, an easily distributable and manageable antigen capture enzyme-linked immunosorbent assay (ELISA) for the detection of orthopoxviruses was developed to facilitate particle detection. By comparing the virus particle binding properties of polyclonal antibodies developed against surface-exposed attachment or fusion proteins, the surface protein A27 was found to be a well-bound, highly immunogenic and exposed target for antibodies aiming at virus particle detection. Subsequently, eight monoclonal anti-A27 antibodies were generated and characterized by peptide epitope mapping and surface plasmon resonance measurements. All antibodies were found to bind with high affinity to two epitopes at the heparin binding site of A27, toward either the N- or C-terminal of the crucial KKEP-segment of A27. Two antibodies recognizing different epitopes were implemented in an antigen capture ELISA. Validation showed robust detection of virus particles from 11 different orthopoxvirus isolates pathogenic to humans, with the exception of MVA, which is apathogenic to humans. Most orthopoxviruses could be detected reliably for viral loads above 1 × 103 PFU/mL. To our knowledge, this is the first solely monoclonal and therefore reproducible antibody-based antigen capture ELISA able to detect all human pathogenic orthopoxviruses including monkeypox virus, except variola virus which was not included. Therefore, the newly developed antibody-based assay represents important progress towards feasible particle detection of this important genus of viruses.
Collapse
Affiliation(s)
- Daniel Stern
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Diana Pauly
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Martin Zydek
- Biosystems Technology, Institute of Applied Life Sciences, Technical University of Applied Sciences, Wildau, Germany
| | - Lilija Miller
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Janett Piesker
- Advanced Light and Electron Microscopy (ZBS 4), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Michael Laue
- Advanced Light and Electron Microscopy (ZBS 4), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Fred Lisdat
- Biosystems Technology, Institute of Applied Life Sciences, Technical University of Applied Sciences, Wildau, Germany
| | - Martin B. Dorner
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Brigitte G. Dorner
- Biological Toxins (ZBS 3), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Andreas Nitsche
- Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
12
|
Liu Q, Fan C, Zhou S, Guo Y, Zuo Q, Ma J, Liu S, Wu X, Peng Z, Fan T, Guo C, Shen Y, Huang W, Li B, He Z, Wang Y. Bioluminescent imaging of vaccinia virus infection in immunocompetent and immunodeficient rats as a model for human smallpox. Sci Rep 2015; 5:11397. [PMID: 26235050 PMCID: PMC4522659 DOI: 10.1038/srep11397] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/11/2015] [Indexed: 01/23/2023] Open
Abstract
Due to the increasing concern of using smallpox virus as biological weapons for terrorist attack, there is renewed interest in studying the pathogenesis of human smallpox and development of new therapies. Animal models are highly demanded for efficacy and safety examination of new vaccines and therapeutic drugs. Here, we demonstrated that both wild type and immunodeficient rats infected with an engineered vaccinia virus carrying Firefly luciferase reporter gene (rTV-Fluc) could recapitulate infectious and clinical features of human smallpox. Vaccinia viral infection in wild type Sprague-Dawley (SD) rats displayed a diffusible pattern in various organs, including liver, head and limbs. The intensity of bioluminescence generated from rTV-Fluc correlated well with viral loads in tissues. Moreover, neutralizing antibodies had a protective effect against virus reinfection. The recombination activating gene 2 (Rag2) knockout rats generated by transcription activator-like effector nucleases (TALENs) technology were further used to examine the infectivity of the rTV-Fluc in immunodeficient populations. Here we demonstrated that Rag2-/- rats were more susceptible to rTV-Fluc than SD rats with a slower virus clearance rate. Therefore, the rTV-Fluc/SD rats and rTV-Fluc/Rag2-/- rats are suitable visualization models, which recapitulate wild type or immunodeficient populations respectively, for testing human smallpox vaccine and antiviral drugs.
Collapse
Affiliation(s)
- Qiang Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Changfa Fan
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Shuya Zhou
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Yanan Guo
- Biocytogen Co., Ltd, Beijing, 101111, China
| | - Qin Zuo
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Jian Ma
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Susu Liu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Xi Wu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Zexu Peng
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Tao Fan
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | | | | | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Baowen Li
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Zhengming He
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, 100050, China
| |
Collapse
|
13
|
Replicating poxviruses for human cancer therapy. J Microbiol 2015; 53:209-18. [PMID: 25845536 DOI: 10.1007/s12275-015-5041-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/04/2015] [Accepted: 03/19/2015] [Indexed: 01/29/2023]
Abstract
Naturally occurring oncolytic viruses are live, replication-proficient viruses that specifically infect human cancer cells while sparing normal cell counterparts. Since the eradication of smallpox in the 1970s with the aid of vaccinia viruses, the vaccinia viruses and other genera of poxviruses have shown various degrees of safety and efficacy in pre-clinical or clinical application for human anti-cancer therapeutics. Furthermore, we have recently discovered that cellular tumor suppressor genes are important in determining poxviral oncolytic tropism. Since carcinogenesis is a multi-step process involving accumulation of both oncogene and tumor suppressor gene abnormalities, it is interesting that poxvirus can exploit abnormal cellular tumor suppressor signaling for its oncolytic specificity and efficacy. Many tumor suppressor genes such as p53, ATM, and RB are known to play important roles in genomic fidelity/maintenance. Thus, tumor suppressor gene abnormality could affect host genomic integrity and likely disrupt intact antiviral networks due to accumulation of genetic defects, which would in turn result in oncolytic virus susceptibility. This review outlines the characteristics of oncolytic poxvirus strains, including vaccinia, myxoma, and squirrelpox virus, recent progress in elucidating the molecular connection between oncogene/tumor suppressor gene abnormalities and poxviral oncolytic tropism, and the associated preclinical/clinical implications. I would also like to propose future directions in the utility of poxviruses for oncolytic virotherapy.
Collapse
|
14
|
Postchallenge administration of brincidofovir protects healthy and immune-deficient mice reconstituted with limited numbers of T cells from lethal challenge with IHD-J-Luc vaccinia virus. J Virol 2015; 89:3295-307. [PMID: 25589648 DOI: 10.1128/jvi.03340-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Protection from lethality by postchallenge administration of brincidofovir (BCV, CMX001) was studied in normal and immune-deficient (nude, nu/nu) BALB/c mice infected with vaccinia virus (VACV). Whole-body bioluminescence imaging was used to record total fluxes in the nasal cavity, lungs, spleen, and liver and to enumerate pox lesions on tails of mice infected via the intranasal route with 10(5) PFU of recombinant IHD-J-Luc VACV expressing luciferase. Areas under the flux curve (AUCs) were calculated for individual mice to assess viral loads. A three-dose regimen of 20 mg/kg BCV administered every 48 h starting either on day 1 or day 2 postchallenge protected 100% of mice. Initiating BCV treatment earlier was more efficient in reducing viral loads and in providing protection from pox lesion development. All BCV-treated mice that survived challenge were also protected from rechallenge with IHD-J-Luc or WRvFire VACV without additional treatment. In immune-deficient mice, BCV protected animals from lethality and reduced viral loads while animals were on the drug. Viral recrudescence occurred within 4 to 9 days, and mice succumbed ∼10 to 20 days after treatment termination. Nude mice reconstituted with 10(5) T cells prior to challenge with 10(4) PFU of IHD-J-Luc and treated with BCV postchallenge survived the infection, cleared the virus from all organs, and survived rechallenge with 10(5) PFU of IHD-J-Luc VACV without additional BCV treatment. Together, these data suggest that BCV protects immunocompetent and partially T cell-reconstituted immune-deficient mice from lethality, reduces viral dissemination in organs, prevents pox lesion development, and permits generation of VACV-specific memory. IMPORTANCE Mass vaccination is the primary element of the public health response to a smallpox outbreak. In addition to vaccination, however, antiviral drugs are required for individuals with uncertain exposure status to smallpox or for whom vaccination is contraindicated. Whole-body bioluminescence imaging was used to study the effect of brincidofovir (BCV) in normal and immune-deficient (nu/nu) mice infected with vaccinia virus, a model of smallpox. Postchallenge administration of 20 mg/kg BCV rescued normal and immune-deficient mice partially reconstituted with T cells from lethality and significantly reduced viral loads in organs. All BCV-treated mice that survived infection were protected from rechallenge without additional treatment. In immune-deficient mice, BCV extended survival. The data show that BCV controls viral replication at the site of challenge and reduces viral dissemination to internal organs, thus providing a shield for the developing adaptive immunity that clears the host of virus and builds virus-specific immunological memory.
Collapse
|
15
|
Hagen CJ, Titong A, Sarnoski EA, Verardi PH. Antibiotic-dependent expression of early transcription factor subunits leads to stringent control of vaccinia virus replication. Virus Res 2014; 181:43-52. [PMID: 24394294 DOI: 10.1016/j.virusres.2013.12.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 12/12/2013] [Accepted: 12/17/2013] [Indexed: 12/31/2022]
Abstract
The use of vaccinia virus (VACV) as the vaccine against variola virus resulted in the eradication of smallpox. VACV has since been used in the development of recombinant vaccine and therapeutic vectors, but complications associated with uncontrolled viral replication have constrained its use as a live viral vector. We propose to improve the safety of VACV as a live-replicating vector by using elements of the tet operon to control the transcription of genes that are essential for viral growth. Poxviruses encode all enzymes and factors necessary for their replication within the host cell cytoplasm. One essential VACV factor is the vaccinia early transcription factor (VETF) packaged into the viral core. This heterodimeric protein is required for expression of early VACV genes. VETF is composed of a large subunit encoded by the A7L gene and a small subunit encoded by the D6R gene. Two recombinant VACVs were generated in which either the A7L or D6R gene was placed under the control of tet operon elements to allow their transcription, and therefore viral replication, to be dependent on tetracycline antibiotics such as doxycycline. In the absence of inducers, no plaques were produced but abortively infected cells could be identified by expression of a reporter gene. In the presence of doxycycline, both recombinant viruses replicated indistinguishably from the wild-type strain. This stringent control of VACV replication can be used for the development of safer, next-generation VACV vaccines and therapeutic vectors. Such replication-inducible VACVs would only replicate when administered with tetracycline antibiotics, and if adverse events were to occur, treatment would be as simple as antibiotic cessation.
Collapse
Affiliation(s)
- Caitlin J Hagen
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture and Natural Resources, University of Connecticut, Storrs, CT 06269, United States
| | - Allison Titong
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture and Natural Resources, University of Connecticut, Storrs, CT 06269, United States
| | - Ethan A Sarnoski
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture and Natural Resources, University of Connecticut, Storrs, CT 06269, United States
| | - Paulo H Verardi
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture and Natural Resources, University of Connecticut, Storrs, CT 06269, United States.
| |
Collapse
|
16
|
Bissa M, Pacchioni SM, Zanotto C, De Giuli Morghen C, Illiano E, Granucci F, Zanoni I, Broggi A, Radaelli A. Systemically administered DNA and fowlpox recombinants expressing four vaccinia virus genes although immunogenic do not protect mice against the highly pathogenic IHD-J vaccinia strain. Virus Res 2013; 178:374-82. [PMID: 24050999 PMCID: PMC9533858 DOI: 10.1016/j.virusres.2013.09.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/05/2013] [Accepted: 09/09/2013] [Indexed: 11/30/2022]
Abstract
The first-generation smallpox vaccine was based on live vaccinia virus (VV) and it successfully eradicated the disease worldwide. Therefore, it was not administered any more after 1980, as smallpox no longer existed as a natural infection. However, emerging threats by terrorist organisations has prompted new programmes for second-generation vaccine development based on attenuated VV strains, which have been shown to cause rare but serious adverse events in immunocompromised patients. Considering the closely related animal poxviruses that might also be used as bioweapons, and the increasing number of unvaccinated young people and AIDS-affected immunocompromised subjects, a safer and more effective smallpox vaccine is still required. New avipoxvirus-based vectors should improve the safety of conventional vaccines, and protect from newly emerging zoonotic orthopoxvirus diseases and from the threat of deliberate release of variola or monkeypox virus in a bioterrorist attack. In this study, DNA and fowlpox recombinants expressing the L1R, A27L, A33R and B5R genes were constructed and evaluated in a pre-clinical trial in mouse, following six prime/boost immunisation regimens, to compare their immunogenicity and protective efficacy against a challenge with the lethal VV IHD-J strain. Although higher numbers of VV-specific IFNγ-producing T lymphocytes were observed in the protected mice, the cytotoxic T-lymphocyte response and the presence of neutralising antibodies did not always correlate with protection. In spite of previous successful results in mice, rabbits and monkeys, where SIV/HIV transgenes were expressed by the fowlpox vector, the immune response elicited by these recombinants was low, and most of the mice were not protected.
Collapse
Affiliation(s)
- Massimiliano Bissa
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Safety mechanism assisted by the repressor of tetracycline (SMART) vaccinia virus vectors for vaccines and therapeutics. Proc Natl Acad Sci U S A 2013; 110:15407-12. [PMID: 23988330 DOI: 10.1073/pnas.1314483110] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Replication-competent viruses, such as Vaccinia virus (VACV), are powerful tools for the development of oncolytic viral therapies and elicit superior immune responses when used as vaccine and immunotherapeutic vectors. However, severe complications from uncontrolled viral replication can occur, particularly in immunocompromised individuals or in those with other predisposing conditions. VACVs constitutively expressing interferon-γ (IFN-γ) replicate in cell culture indistinguishably from control viruses; however, they replicate in vivo to low or undetectable levels, and are rapidly cleared even in immunodeficient animals. In an effort to develop safe and highly effective replication-competent VACV vectors, we established a system to inducibly express IFN-γ. Our SMART (safety mechanism assisted by the repressor of tetracycline) vectors are designed to express the tetracycline repressor under a constitutive VACV promoter and IFN-γ under engineered tetracycline-inducible promoters. Immunodeficient SCID mice inoculated with VACVs not expressing IFN-γ demonstrated severe weight loss, whereas those given VACVs expressing IFN-γ under constitutive VACV promoters showed no signs of infection. Most importantly, mice inoculated with a VACV expressing the IFN-γ gene under an inducible promoter remained healthy in the presence of doxycycline, but exhibited severe weight loss in the absence of doxycycline. In this study, we developed a safety mechanism for VACV based on the conditional expression of IFN-γ under a tightly controlled tetracycline-inducible VACV promoter for use in vaccines and oncolytic cancer therapies.
Collapse
|
18
|
Golden JW, Josleyn M, Mucker EM, Hung CF, Loudon PT, Wu TC, Hooper JW. Side-by-side comparison of gene-based smallpox vaccine with MVA in nonhuman primates. PLoS One 2012; 7:e42353. [PMID: 22860117 PMCID: PMC3409187 DOI: 10.1371/journal.pone.0042353] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 07/04/2012] [Indexed: 11/25/2022] Open
Abstract
Orthopoxviruses remain a threat as biological weapons and zoonoses. The licensed live-virus vaccine is associated with serious health risks, making its general usage unacceptable. Attenuated vaccines are being developed as alternatives, the most advanced of which is modified-vaccinia virus Ankara (MVA). We previously developed a gene-based vaccine, termed 4pox, which targets four orthopoxvirus antigens, A33, B5, A27 and L1. This vaccine protects mice and non-human primates from lethal orthopoxvirus disease. Here, we investigated the capacity of the molecular adjuvants GM-CSF and Escherichia coli heat-labile enterotoxin (LT) to enhance the efficacy of the 4pox gene-based vaccine. Both adjuvants significantly increased protective antibody responses in mice. We directly compared the 4pox plus LT vaccine against MVA in a monkeypox virus (MPXV) nonhuman primate (NHP) challenge model. NHPs were vaccinated twice with MVA by intramuscular injection or the 4pox/LT vaccine delivered using a disposable gene gun device. As a positive control, one NHP was vaccinated with ACAM2000. NHPs vaccinated with each vaccine developed anti-orthopoxvirus antibody responses, including those against the 4pox antigens. After MPXV intravenous challenge, all control NHPs developed severe disease, while the ACAM2000 vaccinated animal was well protected. All NHPs vaccinated with MVA were protected from lethality, but three of five developed severe disease and all animals shed virus. All five NHPs vaccinated with 4pox/LT survived and only one developed severe disease. None of the 4pox/LT-vaccinated animals shed virus. Our findings show, for the first time, that a subunit orthopoxvirus vaccine delivered by the same schedule can provide a degree of protection at least as high as that of MVA.
Collapse
Affiliation(s)
- Joseph W. Golden
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Matthew Josleyn
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Eric M. Mucker
- Department of Viral Therapeutics, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Peter T. Loudon
- Pfizer, Sandwich Laboratories, Sandwich, Kent, United Kingdom
| | - T. C. Wu
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Jay W. Hooper
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
19
|
Crowley CR, Solomons NW, Schümann K. Targeted provision of oral iron: the evolution of a practical screening option. Adv Nutr 2012; 3:560-9. [PMID: 22797993 PMCID: PMC3649727 DOI: 10.3945/an.111.001149] [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] [Indexed: 01/08/2023] Open
Abstract
Universal oral iron supplementation, undertaken according to 1998 WHO guidelines, produced adverse consequences among some children in malaria-endemic areas. Prompted by the Pemba trial, which revealed excessive hospitalizations and deaths, WHO advised that iron supplementation in such regions be accompanied by previous screening for iron deficiency. This agenda, however, poses issues of cost, benefit, acceptability, technical feasibility, and reliability of such screening. The cost of equipment and personnel is balanced against savings from iron supplements spared and treatment for morbidity averted. Costs aside, the most efficacious acceptable screening approach for avoiding hospitalization and deaths must be fielded. Screening before supplementation can be used to assess hematological, iron, and possible inflammatory status to differentiate the source of decreased hemoglobin concentration. Iron deficiency has often been inferred from hematological status markers. The need for extraction of blood, albeit capillary in origin, and high assay costs limit the use of validated methods in screening. Noninvasive methods, i.e., not requiring the extraction of blood, provide the most acceptable and potentially least expensive approach for determining hematological or iron status. Although a noninvasive technique for iron and inflammatory status would be the ideal, it is unattained. Field-friendly, skin-probe hemoglobin devices, derived from instruments for clinical settings, are being developed and tested for eventual rollout in malarial areas. Given a firm grounding for the theoretical requirements needed to advance the screening agenda, evaluation and monitoring of the performance of screening devices can proceed hand in hand.
Collapse
Affiliation(s)
- Caitlin R. Crowley
- Center for Studies of Sensory Impairment, Aging and Metabolism (CeSSIAM), Guatemala City, Guatemala; and
| | - Noel W. Solomons
- Center for Studies of Sensory Impairment, Aging and Metabolism (CeSSIAM), Guatemala City, Guatemala; and,To whom correspondence should be addressed. E-mail:
| | - Klaus Schümann
- Research Center for Nutrition and Food Science, Center for Diet and Disease, Technische Universität München, Freising, Germany
| |
Collapse
|
20
|
Golden JW, Hooper JW. The strategic use of novel smallpox vaccines in the post-eradication world. Expert Rev Vaccines 2012; 10:1021-35. [PMID: 21806397 PMCID: PMC9491137 DOI: 10.1586/erv.11.46] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We still face a threat of orthopoxviruses in the form of biological weapons and emerging zoonoses. Therefore, there is a need to maintain a comprehensive defense strategy to counter the low-probability, high-impact threat of smallpox, as well as the ongoing threat of naturally occurring orthopoxvirus disease. The currently licensed live-virus smallpox vaccine ACAM2000 is effective, but associated with serious and even life-threatening adverse events. The health threat posed by this vaccine, and other previously licensed vaccines, has prevented many first responders, and even many in the military, from receiving a vaccine against smallpox. At the same time, global immunity produced during the smallpox eradication campaign is waning. Here, we review novel subunit/component vaccines and how they might play roles in unconventional strategies to defend against emerging orthopoxvirus diseases throughout the world and against smallpox used as a weapon of mass destruction.
Collapse
Affiliation(s)
- Joseph W Golden
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | | |
Collapse
|
21
|
Vliegen I, Yang G, Hruby D, Jordan R, Neyts J. Deletion of the vaccinia virus F13L gene results in a highly attenuated virus that mounts a protective immune response against subsequent vaccinia virus challenge. Antiviral Res 2011; 93:160-6. [PMID: 22138484 DOI: 10.1016/j.antiviral.2011.11.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 11/10/2011] [Accepted: 11/16/2011] [Indexed: 11/17/2022]
Abstract
Vaccinia virus F13L encodes the envelope protein p37, which is the target of the anti-pox virus drug ST-246 (Yang et al., 2005) and that is required for production of extracellular vaccinia virus. The F13L (p37)-deleted (and ST-246 resistant) vaccinia virus recombinant (Vac-ΔF13L) produced smaller plaques than the wild-type vaccinia (Western Reserve vaccinia). In addition, Vac-ΔF13L proved, when inoculated either intravenously or intracutaneously in both immunocompetent and immunodeficient (athymic nude or SCID) mice, to be severely attenuated. Intravenous or intracutaneous inoculation of immunocompetent mice with the ΔF13L virus efficiently protected against a subsequent intravenous, intracutaneous or intranasal challenge with vaccinia WR (Western Reserve). This was corroborated by the observation that Vac-ΔF13L induced a humoral immune response against vaccinia following either intravenous or intracutaneous challenge. In conclusion, F13L-deleted vaccinia virus may have the potential to be developed as a smallpox vaccine.
Collapse
Affiliation(s)
- Inge Vliegen
- Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KULeuven, Leuven, Belgium
| | | | | | | | | |
Collapse
|
22
|
Golden JW, Zaitseva M, Kapnick S, Fisher RW, Mikolajczyk MG, Ballantyne J, Golding H, Hooper JW. Polyclonal antibody cocktails generated using DNA vaccine technology protect in murine models of orthopoxvirus disease. Virol J 2011; 8:441. [PMID: 21933385 PMCID: PMC3192780 DOI: 10.1186/1743-422x-8-441] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Accepted: 09/20/2011] [Indexed: 12/17/2022] Open
Abstract
Background Previously we demonstrated that DNA vaccination of nonhuman primates (NHP) with a small subset of vaccinia virus (VACV) immunogens (L1, A27, A33, B5) protects against lethal monkeypox virus challenge. The L1 and A27 components of this vaccine target the mature virion (MV) whereas A33 and B5 target the enveloped virion (EV). Results Here, we demonstrated that the antibodies produced in vaccinated NHPs were sufficient to confer protection in a murine model of lethal Orthopoxvirus infection. We further explored the concept of using DNA vaccine technology to produce immunogen-specific polyclonal antibodies that could then be combined into cocktails as potential immunoprophylactic/therapeutics. Specifically, we used DNA vaccines delivered by muscle electroporation to produce polyclonal antibodies against the L1, A27, A33, and B5 in New Zealand white rabbits. The polyclonal antibodies neutralized both MV and EV in cell culture. The ability of antibody cocktails consisting of anti-MV, anti-EV, or a combination of anti-MV/EV to protect BALB/c mice was evaluated as was the efficacy of the anti-MV/EV mixture in a mouse model of progressive vaccinia. In addition to evaluating weight loss and lethality, bioimaging technology was used to characterize the spread of the VACV infections in mice. We found that the anti-EV cocktail, but not the anti-MV cocktail, limited virus spread and lethality. Conclusions A combination of anti-MV/EV antibodies was significantly more protective than anti-EV antibodies alone. These data suggest that DNA vaccine technology could be used to produce a polyclonal antibody cocktail as a possible product to replace vaccinia immune globulin.
Collapse
Affiliation(s)
- Joseph W Golden
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Sato H. Countermeasures and vaccination against terrorism using smallpox: pre-event and post-event smallpox vaccination and its contraindications. Environ Health Prev Med 2011; 16:281-9. [PMID: 21431786 PMCID: PMC3156838 DOI: 10.1007/s12199-010-0200-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 11/16/2010] [Indexed: 11/26/2022] Open
Abstract
Smallpox, when used as a biological weapon, presents a serious threat to civilian populations. Core components of the public health management of a terrorism attack using smallpox are: vaccination (ring vaccination and mass vaccination), adverse event monitoring, confirmed and suspected smallpox case management, contact management, identifying, tracing, monitoring contacts, and quarantine. Above all, pre-event and post-event vaccination is an indispensable part of the strategies. Since smallpox patients are most infectious from onset of the rash through the first 7-10 days of the rash, vaccination should be administered promptly within a limited time frame. However, vaccination can accompany complications, such as postvaccinial encephalitis, progressive vaccinia, eczema vaccinatum, and generalized vaccinia. Therefore, vaccination is not recommended for certain groups. Public health professionals, as well as physicians and government officials, should also be well equipped with all information necessary for appropriate and effective smallpox management in the face of such a bioterrorism attack.
Collapse
Affiliation(s)
- Hajime Sato
- Department of Public Health, Graduate School of Medicine, The University of Tokyo, Hongo, Bunkyo-ku, Japan.
| |
Collapse
|
24
|
Evaluating the orthopoxvirus type I interferon-binding molecule as a vaccine target in the vaccinia virus intranasal murine challenge model. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1656-65. [PMID: 20844086 DOI: 10.1128/cvi.00235-10] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The biological threat imposed by orthopoxviruses warrants the development of safe and effective vaccines. We developed a candidate orthopoxvirus DNA-based vaccine, termed 4pox, which targets four viral structural components, A33, B5, A27, and L1. While this vaccine protects mice and nonhuman primates from lethal infections, we are interested in further enhancing its potency. One approach to enhance potency is to include additional orthopoxvirus immunogens. Here, we investigated whether vaccination with the vaccinia virus (VACV) interferon (IFN)-binding molecule (IBM) could protect BALB/c mice against lethal VACV challenge. We found that vaccination with this molecule failed to significantly protect mice from VACV when delivered alone. IBM modestly augmented protection when delivered together with the 4pox vaccine. All animals receiving the 4pox vaccine plus IBM lived, whereas only 70% of those receiving a single dose of 4pox vaccine survived. Mapping studies using truncated mutants revealed that vaccine-generated antibodies spanned the immunoglobulin superfamily domains 1 and 2 and, to a lesser extent, 3 of the IBM. These antibodies inhibited IBM cell binding and IFN neutralization activity, indicating that they were functionally active. This study shows that DNA vaccination with the VACV IBM results in a robust immune response but that this response does not significantly enhance protection in a high-dose challenge model.
Collapse
|
25
|
Direct TLR2 signaling is critical for NK cell activation and function in response to vaccinia viral infection. PLoS Pathog 2010; 6:e1000811. [PMID: 20300608 PMCID: PMC2837413 DOI: 10.1371/journal.ppat.1000811] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Accepted: 02/05/2010] [Indexed: 11/19/2022] Open
Abstract
Natural killer (NK) cells play an essential role in innate immune control of poxviral infections in vivo. However, the mechanism(s) underlying NK cell activation and function in response to poxviruses remains poorly understood. In a mouse model of infection with vaccinia virus (VV), the most studied member of the poxvirus family, we identified that the Toll-like receptor (TLR) 2-myeloid differentiating factor 88 (MyD88) pathway was critical for the activation of NK cells and the control of VV infection in vivo. We further showed that TLR2 signaling on NK cells, but not on accessory cells such as dendritic cells (DCs), was necessary for NK cell activation and that this intrinsic TLR2-MyD88 signaling pathway was required for NK cell activation and played a critical role in the control of VV infection in vivo. In addition, we showed that the activating receptor NKG2D was also important for efficient NK activation and function, as well as recognition of VV-infected targets. We further demonstrated that VV could directly activate NK cells via TLR2 in the presence of cytokines in vitro and TLR2-MyD88-dependent activation of NK cells by VV was mediated through the phosphatidylinositol 3-kinase (PI3K)-extracellular signal-regulated kinase (ERK) pathway. Taken together, these results represent the first evidence that intrinsic TLR signaling is critical for NK cell activation and function in the control of a viral infection in vivo, indicate that multiple pathways are required for efficient NK cell activation and function in response to VV infection, and may provide important insights into the design of effective strategies to combat poxviral infections. NK cells are an important component of innate immunity in fighting against poxviral infections in vivo. However, how NK cells are activated and exert their function in controlling poxviruses remains poorly understood. In this paper, we found that VV, the most studied member of the poxvirus family, could directly activate TLR2 on NK cells and that the direct TLR2 stimulation was critical for NK cell activation and function in the control of VV infection in vivo. We further showed that TLR2-dependent NK cell activation by VV was mediated through the PI3K-ERK pathway. In addition, we demonstrated that the activating receptor NKG2D was also required for efficient NK cell activation and function. Collectively, these results represent the first evidence that direct TLR signaling is crucial to NK cell activation and function in the control of a viral infection in vivo, indicate that multiple pathways are required for efficient NK cell activation, and may provide important insights into the design of effective strategies to combat poxviral infections.
Collapse
|
26
|
Hooper JW, Ferro AM, Golden JW, Silvera P, Dudek J, Alterson K, Custer M, Rivers B, Morris J, Owens G, Smith JF, Kamrud KI. Molecular smallpox vaccine delivered by alphavirus replicons elicits protective immunity in mice and non-human primates. Vaccine 2009; 28:494-511. [PMID: 19833247 DOI: 10.1016/j.vaccine.2009.09.133] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 09/18/2009] [Accepted: 09/30/2009] [Indexed: 11/27/2022]
Abstract
Naturally occurring smallpox was eradicated as a result of successful vaccination campaigns during the 1960s and 1970s. Because of its highly contagious nature and high mortality rate, smallpox has significant potential as a biological weapon. Unfortunately, the current vaccine for orthopoxviruses is contraindicated for large portions of the population. Thus, there is a need for new, safe, and effective orthopoxvirus vaccines. Alphavirus replicon vectors, derived from strains of Venezuelan equine encephalitis virus, are being used to develop alternatives to the current smallpox vaccine. Here, we demonstrated that virus-like replicon particles (VRPs) expressing the vaccinia virus A33R, B5R, A27L, and L1R genes elicited protective immunity in mice comparable to vaccination with live-vaccinia virus. Furthermore, cynomolgus macaques vaccinated with a combination of the four poxvirus VRPs (4pox-VRP) developed antibody responses to each antigen. These antibody responses were able to neutralize and inhibit the spread of both vaccinia virus and monkeypox virus. Macaques vaccinated with 4pox-VRP, flu HA VRP (negative control), or live-vaccinia virus (positive control) were challenged intravenously with 5 x 10(6)pfu of monkeypox virus 1 month after the second VRP vaccination. Four of the six negative control animals succumbed to monkeypox and the remaining two animals demonstrated either severe or grave disease. Importantly, all 10 macaques vaccinated with the 4pox-VRP vaccine survived without developing severe disease. These findings revealed that a single-boost VRP smallpox vaccine shows promise as a safe alternative to the currently licensed live-vaccinia virus smallpox vaccine.
Collapse
Affiliation(s)
- Jay W Hooper
- US Army Medical Research Institute of Infectious Diseases, Virology Division, 1425 Porter Street, Fort Detrick, MD 21702, United States.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Liszewski MK, Leung MK, Hauhart R, Fang CJ, Bertram P, Atkinson JP. Smallpox inhibitor of complement enzymes (SPICE): dissecting functional sites and abrogating activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 183:3150-9. [PMID: 19667083 PMCID: PMC2899487 DOI: 10.4049/jimmunol.0901366] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although smallpox was eradicated as a global illness more than 30 years ago, variola virus and other related pathogenic poxviruses, such as monkeypox, remain potential bioterrorist weapons or could re-emerge as natural infections. Poxviruses express virulence factors that down-modulate the host's immune system. We previously compared functional profiles of the poxviral complement inhibitors of smallpox, vaccinia, and monkeypox known as SPICE, VCP (or VICE), and MOPICE, respectively. SPICE was the most potent regulator of human complement and attached to cells via glycosaminoglycans. The major goals of the present study were to further characterize the complement regulatory and heparin binding sites of SPICE and to evaluate a mAb that abrogates its function. Using substitution mutagenesis, we established that (1) elimination of the three heparin binding sites severely decreases but does not eliminate glycosaminoglycan binding, (2) there is a hierarchy of activity for heparin binding among the three sites, and (3) complement regulatory sites overlap with each of the three heparin binding motifs. By creating chimeras with interchanges of SPICE and VCP residues, a combination of two SPICE amino acids (H77 plus K120) enhances VCP activity approximately 200-fold. Also, SPICE residue L131 is critical for both complement regulatory function and accounts for the electrophoretic differences between SPICE and VCP. An evolutionary history for these structure-function adaptations of SPICE is proposed. Finally, we identified and characterized a mAb that inhibits the complement regulatory activity of SPICE, MOPICE, and VCP and thus could be used as a therapeutic agent.
Collapse
Affiliation(s)
- M. Kathryn Liszewski
- Department of Medicine/Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110
| | - Marilyn K. Leung
- Department of Medicine/Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110
| | - Richard Hauhart
- Department of Medicine/Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110
| | - Celia J. Fang
- Medical Service, Veterans Administration Medical Center, San Francisco, CA 94121
| | - Paula Bertram
- Department of Medicine/Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110
| | - John P. Atkinson
- Department of Medicine/Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
28
|
Damon IK, Davidson WB, Hughes CM, Olson VA, Smith SK, Holman RC, Frey SE, Newman F, Belshe RB, Yan L, Karem K. Evaluation of smallpox vaccines using variola neutralization. J Gen Virol 2009; 90:1962-1966. [PMID: 19339477 PMCID: PMC2887573 DOI: 10.1099/vir.0.010553-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Accepted: 03/28/2009] [Indexed: 11/18/2022] Open
Abstract
The search for a 'third'-generation smallpox vaccine has resulted in the development and characterization of several vaccine candidates. A significant barrier to acceptance is the absence of challenge models showing induction of correlates of protective immunity against variola virus. In this light, virus neutralization provides one of few experimental methods to show specific 'in vitro' activity of vaccines against variola virus. Here, we provide characterization of the ability of a modified vaccinia virus Ankara vaccine to induce variola virus-neutralizing antibodies, and we provide comparison with the neutralization elicited by standard Dryvax vaccination.
Collapse
Affiliation(s)
- Inger K. Damon
- Centers for Disease Control and Prevention, National Center for Zoonotic, Vector-borne, and Enteric Diseases, Division of Viral and Rickettsial Diseases, 1600 Clifton Road NE, Atlanta, GA 30333, USA
| | - Whitni B. Davidson
- Centers for Disease Control and Prevention, National Center for Zoonotic, Vector-borne, and Enteric Diseases, Division of Viral and Rickettsial Diseases, 1600 Clifton Road NE, Atlanta, GA 30333, USA
| | - Christine M. Hughes
- Centers for Disease Control and Prevention, National Center for Zoonotic, Vector-borne, and Enteric Diseases, Division of Viral and Rickettsial Diseases, 1600 Clifton Road NE, Atlanta, GA 30333, USA
| | - Victoria A. Olson
- Centers for Disease Control and Prevention, National Center for Zoonotic, Vector-borne, and Enteric Diseases, Division of Viral and Rickettsial Diseases, 1600 Clifton Road NE, Atlanta, GA 30333, USA
| | - Scott K. Smith
- Centers for Disease Control and Prevention, National Center for Zoonotic, Vector-borne, and Enteric Diseases, Division of Viral and Rickettsial Diseases, 1600 Clifton Road NE, Atlanta, GA 30333, USA
| | - Robert C. Holman
- Centers for Disease Control and Prevention, National Center for Zoonotic, Vector-borne, and Enteric Diseases, Division of Viral and Rickettsial Diseases, 1600 Clifton Road NE, Atlanta, GA 30333, USA
| | - Sharon E. Frey
- Saint Louis University School of Medicine, Division of Infectious Diseases and Immunology, Edward A. Doisy Research Center, 8th Floor, 1100 S. Grand Blvd, St Louis, MO 63104, USA
| | - Frances Newman
- Saint Louis University School of Medicine, Division of Infectious Diseases and Immunology, Edward A. Doisy Research Center, 8th Floor, 1100 S. Grand Blvd, St Louis, MO 63104, USA
| | - Robert B. Belshe
- Saint Louis University School of Medicine, Division of Infectious Diseases and Immunology, Edward A. Doisy Research Center, 8th Floor, 1100 S. Grand Blvd, St Louis, MO 63104, USA
| | - Lihan Yan
- The EMMES Corporation, 401 N. Washington St, Rockville, MD 20850, USA
| | - Kevin Karem
- Centers for Disease Control and Prevention, National Center for Zoonotic, Vector-borne, and Enteric Diseases, Division of Viral and Rickettsial Diseases, 1600 Clifton Road NE, Atlanta, GA 30333, USA
| |
Collapse
|
29
|
|
30
|
Heterogeneity in the A33 protein impacts the cross-protective efficacy of a candidate smallpox DNA vaccine. Virology 2008; 377:19-29. [PMID: 18482742 DOI: 10.1016/j.virol.2008.04.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 04/01/2008] [Accepted: 04/02/2008] [Indexed: 01/05/2023]
Abstract
We previously developed a gene-based vaccine, termed 4pox, which targets four orthopoxvirus proteins (A33, L1, B5, and A27). Because any subunit orthopoxvirus vaccine must protect against multiple species of orthopoxviruses, we are interested in understanding the cross-protective potential of our 4pox vaccine target immunogens. In our current studies, we focused on the A33 immunogen. We found one monoclonal antibody against A33, MAb-1G10, which could not bind the monkeypox virus A33 ortholog, A35. MAb-1G10 binding could be rescued if A35 amino acids 118 and 120 were substituted with those from A33. MAb-1G10 has been shown to protect mice from VACV challenge, thus our findings indicated a protective epitope differs among orthopoxviruses. Accordingly, we tested the cross-protective efficacy of a DNA vaccine consisting of A35R against VACV challenge and compared it to vaccination with A33R DNA. Mice vaccinated with A35R had greater mortality and more weight loss compared to those vaccinated with A33R. These findings demonstrate that despite high homology between A33R orthologs, amino acid differences can impact cross-protection. Furthermore, our results caution that adequate cross-protection by any pan-orthopoxvirus subunit vaccine will require not only careful evaluation of cross-protective immunity, but also of targeting of multiple orthopoxvirus immunogens.
Collapse
|
31
|
Golden JW, Josleyn MD, Hooper JW. Targeting the vaccinia virus L1 protein to the cell surface enhances production of neutralizing antibodies. Vaccine 2008; 26:3507-15. [PMID: 18485547 DOI: 10.1016/j.vaccine.2008.04.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2007] [Revised: 02/22/2008] [Accepted: 04/09/2008] [Indexed: 10/22/2022]
Abstract
The current live-orthopoxvirus vaccine is associated with minor to serious adverse affects, and is contraindicated for use in a significant portion of the population. As an alternative vaccine, we have previously shown that a DNA subunit vaccine (4pox) based on four orthopoxvirus immunogens (L1R, B5R, A27L and A33R) can produce protective immunity against lethal orthopoxvirus challenges in mice and nonhuman primates. Because antibodies are critical for protection against secondary orthopoxvirus infections, we are now interested in strategies that will enhance the humoral immune response against vaccine targets. Here, we tested the immunogenicity of an L1R construct to which a tissue plasminogen activator signal sequence was placed in frame with the full-length L1R gene. The tPA-L1R construct produced a more robust neutralizing antibody response in vaccinated mice when the DNA vaccine was administered by gene-gun as a prime/single boost. When the tPA-L1R construct was substituted for the unmodified L1R gene in the 4pox vaccine, given as a prime and single boost, animals were better protected from lethal challenge with vaccinia virus (VACV). These findings indicate that adding a tPA-leader sequence can enhance the immunogenicity of the L1R gene when given as a DNA vaccine. Furthermore, our results demonstrate that a DNA-based vaccine is capable of establishing protection from lethal orthopoxvirus challenges when administered as a prime and single boost without requiring adjuvant.
Collapse
Affiliation(s)
- Joseph W Golden
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, United States
| | | | | |
Collapse
|
32
|
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
|
33
|
Osborne JD, Da Silva M, Frace AM, Sammons SA, Olsen-Rasmussen M, Upton C, Buller RML, Chen N, Feng Z, Roper RL, Liu J, Pougatcheva S, Chen W, Wohlhueter RM, Esposito JJ. Genomic differences of Vaccinia virus clones from Dryvax smallpox vaccine: the Dryvax-like ACAM2000 and the mouse neurovirulent Clone-3. Vaccine 2007; 25:8807-32. [PMID: 18037545 DOI: 10.1016/j.vaccine.2007.10.040] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Revised: 10/02/2007] [Accepted: 10/08/2007] [Indexed: 10/22/2022]
Abstract
Conventional vaccines used for smallpox eradication were often denoted one or another strain of Vaccinia virus (VACV), even though seed virus was sub-cultured multifariously, which rendered the virion population genetically heterogeneous. ACAM2000 cell culture vaccine, recently licensed in the U.S., consists of a biologically vaccine-like VACV homogeneous-sequence clone from the conventional smallpox vaccine Dryvax, which we verified from Dryvax sequence chromatograms is genetically heterogeneous. ACAM2000 VACV and CL3, a mouse-neurovirulent clone from Dryvax, differ by 572 single nucleotide polymorphisms and 53 insertions-deletions of varied size, including a 4.5-kbp deletion in ACAM2000 and a 6.2-kbp deletion in CL3. The sequence diversity between the two clones precludes precisely defining why CL3 is more pathogenic; however, four genes appear significantly dissimilar to account for virulence differences. CL3 encodes intact immunomodulators interferon-alpha/beta and tumor necrosis factor receptors, which are truncated in ACAM2000. CL3 specifies a Cowpox and Variola virus-like ankyrin-repeat protein that might be associated with proteolysis via ubiquitination. And, CL3 shows an elongated thymidylate kinase, similar to the enzyme of the mouse-neurovirulent VACV-WR, a derivative of the New York City Board of Health vaccine, the origin vaccine of Dryvax. Although ACAM2000 encodes most proteins associated with immunization protection, the cloning probably delimited the variant epitopes and other motifs produced by Dryvax due to its VACV genetic heterogeneity. The sequence information for ACAM2000 and CL3 could be significant for resolving the dynamics of their different proteomes and thereby aid development of safer, more effective vaccines.
Collapse
Affiliation(s)
- John D Osborne
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Preparedness, Detection, and Control of Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Saijo M, Ami Y, Suzaki Y, Nagata N, Iwata N, Hasegawa H, Ogata M, Fukushi S, Mizutani T, Sata T, Kurata T, Kurane I, Morikawa S. LC16m8, a highly attenuated vaccinia virus vaccine lacking expression of the membrane protein B5R, protects monkeys from monkeypox. J Virol 2006; 80:5179-88. [PMID: 16698998 PMCID: PMC1472157 DOI: 10.1128/jvi.02642-05] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2005] [Accepted: 03/06/2006] [Indexed: 11/20/2022] Open
Abstract
The potential threat of smallpox as a bioweapon has led to the production and stockpiling of smallpox vaccine in some countries. Human monkeypox, a rare but important viral zoonosis endemic to central and western Africa, has recently emerged in the United States. Thus, even though smallpox has been eradicated, a vaccinia virus vaccine that can induce protective immunity against smallpox and monkeypox is still invaluable. The ability of the highly attenuated vaccinia virus vaccine strain LC16m8, with a mutation in the important immunogenic membrane protein B5R, to induce protective immunity against monkeypox in nonhuman primates was evaluated in comparison with the parental Lister strain. Monkeys were immunized with LC16m8 or Lister and then infected intranasally or subcutaneously with monkeypox virus strain Liberia or Zr-599, respectively. Immunized monkeys showed no symptoms of monkeypox in the intranasal-inoculation model, while nonimmunized controls showed typical symptoms. In the subcutaneous-inoculation model, monkeys immunized with LC16m8 showed no symptoms of monkeypox except for a mild ulcer at the site of monkeypox virus inoculation, and those immunized with Lister showed no symptoms of monkeypox, while nonimmunized controls showed lethal and typical symptoms. These results indicate that LC16m8 prevents lethal monkeypox in monkeys, and they suggest that LC16m8 may induce protective immunity against smallpox.
Collapse
Affiliation(s)
- Masayuki Saijo
- Special Pathogens Laboratory, Department of Virology 1, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Fry DE. Disaster Planning for Unconventional Acts of Civilian Terrorism. Curr Probl Surg 2006; 43:253-315. [PMID: 16581341 DOI: 10.1067/j.cpsurg.2006.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Donald E Fry
- University of New Mexico School of Medicine, Albuquerque, USA
| |
Collapse
|
36
|
Abstract
The WHO declared smallpox eradicated in 1980. However, concern over its potential use by terrorists or in biowarfare has led to striking growth in research related to this much-feared disease. Modern molecular techniques and new animal models are advancing our understanding of smallpox and its interaction with the host immune system. Rapid progress is likewise being made in smallpox laboratory diagnostics, smallpox vaccines, and antiviral medications. WHO and several nations are developing stockpiles of smallpox vaccine for use in the event the disease is reintroduced. National and international public-health agencies have also drawn up plans to help with early detection of and response to a smallpox outbreak. These plans hinge on physicians' ability to recognise the clinical features of smallpox and to distinguish it from other illnesses characterised by rashes.
Collapse
Affiliation(s)
- Zack S Moore
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, 2015 Uppergate Drive NE, Atlanta, GA 30322, USA.
| | | | | |
Collapse
|
37
|
Greenberg RN, Schosser RH, Plummer EA, Roberts SE, Caldwell MA, Hargis DL, Rudy DW, Evans ME, Hopkins RJ. Urticaria, Exanthems, and Other Benign Dermatologic Reactions to Smallpox Vaccination in Adults. Clin Infect Dis 2004; 38:958-65. [PMID: 15034827 DOI: 10.1086/382360] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2003] [Accepted: 12/01/2003] [Indexed: 11/03/2022] Open
Abstract
A phase 1 smallpox vaccine trial involving 350 adult volunteers was conducted. Of these subjects, 250 were naive to vaccinia virus vaccine (i.e., "vaccinia naive"). Volunteers received a new cell-cultured smallpox vaccine or a live vaccinia virus vaccine. Nine self-limiting rashes (3.6%) were observed in the vaccinia-naive group. None of the vaccinia-experienced patients had a rash. Rashes appeared 6-19 days after vaccination and had 5 different clinical presentations. Five volunteers had urticarial rashes that resolved within 4-15 days, 1 had an exanthem that lasted 20 days, and 1 each presented with folliculitis, contact dermatitis, and erythematous papules found only on the hands and fingers. Volunteers reported pruritus, tingling, and occasional headaches. Relief was obtained with antihistamine and acetaminophen therapy. No volunteer experienced fever or significant discomfort.
Collapse
Affiliation(s)
- Richard N Greenberg
- Department of Medicine, Department of Veterans Affairs Medical Center, Lexington, Kentucky 40536-0084, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|