1
|
Cheng X, Wang Y, Huang B, Bing J, Wang T, Han R, Huo S, Sun S, Zhao L, Shu C, Deng Y, Tan W. Rational mpox vaccine design: immunogenicity and protective effect of individual and multicomponent proteins in mice. Emerg Microbes Infect 2025; 14:2482702. [PMID: 40105863 PMCID: PMC11951338 DOI: 10.1080/22221751.2025.2482702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/20/2025]
Abstract
The 2022 global mpox virus (MPXV) outbreak highlights the urgent need for safer, next-generation vaccines. We compared the immunogenicity and protective efficacy of individual and multicomponent membrane proteins of MPXV virions in mice to inform the development of a recombinant subunit vaccine against mpox. BALB/c mice were immunized with eukaryotically expressed A35R, A29L, B6R, and M1R proteins, administered individually or in multicomponent combinations with an Al(OH)3 + CpG oligodeoxynucleotide adjuvant. Three multicomponent protein vaccines (A29/B6, A29/B6/M1, and A29/B6/M1/A35) provided complete protection, but others (individual protein and A35/M1 combinations) provided partial protection against challenge with high-lethal doses of vaccinia virus Western Reserve (VACV-WR). Additionally, A29/B6 conferred partial protection, whereas A29/B6/M1 and A29/B6/M1/A35 provided complete protection against ectromelia virus (ECTV), with A29/B6/M1 being most effective. All vaccines induced strong antigen-specific immunoglobulin G (IgG) and cellular immunity, whereas only four (M1, A35/M1, A29/B6/M1, A29/B6/M1/A35) exhibited significant neutralizing activity against MPXV, VACV-Tiantan, and ECTV. Correlation analysis suggested that neutralizing antibodies and A35-/A29-/B6-specific cellular immunity act as complementary defense mechanisms, potentially providing first- and second-line protection against MPXV and related orthopoxviruses. Collectively, A29/B6/M1 demonstrated the best protective efficacy. This study provides novel insights into immunogen optimization and potential mechanisms for the development of vaccines against MPXV and other orthopoxviruses.
Collapse
MESH Headings
- Animals
- Mice, Inbred BALB C
- Mice
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Female
- Ectromelia virus/immunology
- Ectromelia virus/genetics
- Ectromelia, Infectious/prevention & control
- Ectromelia, Infectious/immunology
- Ectromelia, Infectious/virology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Immunogenicity, Vaccine
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Adjuvants, Immunologic/administration & dosage
- Immunity, Cellular
- Oligodeoxyribonucleotides
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
Collapse
Affiliation(s)
- Xueting Cheng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| | - Yawei Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| | - Baoying Huang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| | - Jialuo Bing
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| | - Tangqi Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| | - Ruiwen Han
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| | - Shuting Huo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| | - Shucai Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| | - Li Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| | - Chang Shu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| | - Yao Deng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| | - Wenjie Tan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, People’s Republic of China
| |
Collapse
|
2
|
Shafaati M, Forghani S, Shahsavand Davoudi A, Samiee R, Mohammadi K, Akbarpour S, Seifi A, Salehi M, Zare M. Current advances and challenges in mpox vaccine development: a global landscape. Ther Adv Vaccines Immunother 2025; 13:25151355251314339. [PMID: 39872308 PMCID: PMC11770767 DOI: 10.1177/25151355251314339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
Given the surge in mpox outbreaks in 2022 and the advancements in domestic and international vaccine research, the effectiveness of smallpox vaccines in providing cross-protection against mpox remains crucial. Having learned from the COVID-19 pandemic, it is significant to continue evaluating existing vaccines to ensure their safety and efficacy. Developing new vaccines for widespread use against mpox and its emerging strains also serves as a preventive strategy in the ongoing battle against this dynamic infection. Here's an opportunity to control human-to-human transmission, give short deadlines, and avoid vaccine disparity. Public health systems must take decisive action to prevent the global spread of mpox, particularly among vulnerable groups. This action should include strengthening global surveillance, improving vaccine access, and ensuring equitable distribution, particularly in resource-poor settings, to prevent future outbreaks. This review aims to assess recent advancements and barriers in mpox vaccine development, emphasizing cross-protection and equitable vaccine distribution in resource-poor settings.
Collapse
Affiliation(s)
- Maryam Shafaati
- Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Infectious Diseases Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Shayan Forghani
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Reza Samiee
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Keyhan Mohammadi
- Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Infectious Diseases Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Department of Clinical Pharmacy, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Samaneh Akbarpour
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Occupational Sleep Research Center, Baharloo Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Seifi
- Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Infectious Diseases Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Salehi
- Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Infectious Diseases Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Zare
- Virology Department of Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
3
|
Hicks B, Jones S, Callaby H, Bailey D, Gordon C, Rampling T, Houlihan C, Linley E, Tonge S, Oeser C, Jones R, Pond M, Mehta R, Wright D, Hallis B, Rowe C, Otter A. Evaluation of a multiplexed immunoassay for assessing long-term humoral immunity Orthopoxviruses. Vaccine 2024; 42:126453. [PMID: 39426286 DOI: 10.1016/j.vaccine.2024.126453] [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: 08/17/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The 2022 Monkeypox virus (MPXV) global outbreak boosted development of multiple serological assays to aid understanding of Mpox immunology. OBJECTIVES The study aimed to assess a multiplexed solid-phase electrochemiluminescence immunoassay (Meso Scale Discovery (MSD)) for simultaneous detection of antibodies against MPXV, including A35, E8 and M1 antigens, along with corresponding Vaccina Virus (VACV) homologues and demonstrate its accuracy in assessing antibody titres post-vaccination and infection. METHODS Assay performance was assessed for simultaneous detection of antibodies against MPXV and corresponding VACV antigens. Sensitivity and specificity were evaluated with paediatric negatives (n = 215), pre- and post-IMVANEX vaccinated (n = 80), and MPXV (Clade IIb, n = 39) infected serum samples. RESULTS The assay demonstrated high specificity (75.68 % (CI: 69.01-81.29) - 95.98 % (CI:92.54-97.87)) and sensitivity (62.11 % (CI:52.06-71.21) - 98.59 % (CI:92.44 %-99.93 %)) depending on the Orthopoxvirus antigen. Preferential binding was observed between MPXV-infected individuals and MPXV antigens, while vaccinated individuals exhibited increased binding to VACV antigens. These results highlight differential binding patterns between antigen homologues in related viruses. CONCLUSION Overall, this assay demonstrates high sensitivities in detecting antibodies for multiple relevant MPXV and VACV antigens post-infection and post-vaccination, indicating its utility in understanding immune responses to Orthopoxviruses in current and future outbreaks and evaluating the immunogenicity of new-generation Mpox-specific vaccinations.
Collapse
Affiliation(s)
- Bethany Hicks
- Emerging Pathogen Serology Group, Vaccine Development Evaluation and Preparedness Centre, UK Health Security Agency, Porton Down, Wiltshire, UK.
| | - Scott Jones
- Emerging Pathogen Serology Group, Vaccine Development Evaluation and Preparedness Centre, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Helen Callaby
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK; The Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Daniel Bailey
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Claire Gordon
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Tommy Rampling
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Catherine Houlihan
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Ezra Linley
- Seroepidemiology Unit, UK Health Security Agency, Manchester, UK
| | - Simon Tonge
- Seroepidemiology Unit, UK Health Security Agency, Manchester, UK
| | - Clarissa Oeser
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, Colindale, London, UK
| | - Rachael Jones
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Marcus Pond
- Imperial College Healthcare NHS Trust, London, UK
| | - Ravi Mehta
- Imperial College Healthcare NHS Trust, London, UK
| | - Deborah Wright
- Research and Development, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Bassam Hallis
- Emerging Pathogen Serology Group, Vaccine Development Evaluation and Preparedness Centre, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Cathy Rowe
- Emerging Pathogen Serology Group, Vaccine Development Evaluation and Preparedness Centre, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Ashley Otter
- Emerging Pathogen Serology Group, Vaccine Development Evaluation and Preparedness Centre, UK Health Security Agency, Porton Down, Wiltshire, UK
| |
Collapse
|
4
|
Liang CY, Chao TL, Chao CS, Liu WD, Cheng YC, Chang SY, Chang SC. Monkeypox virus A29L protein as the target for specific diagnosis and serological analysis. Appl Microbiol Biotechnol 2024; 108:522. [PMID: 39570405 PMCID: PMC11582270 DOI: 10.1007/s00253-024-13361-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/30/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
The unexpected monkeypox (Mpox) outbreak has been reported in many non-endemic countries and regions since May 2022. The mutant strains of Mpox virus (MPXV) were found with higher infectivity and greater capability for sustained human-to-human transmission, posing a significant public health threat. MPXV A29L, a protein homolog of vaccinia virus (VACV) A27L, plays an important role in viral attachment to host cell membranes. Therefore, MPXV A29L is considered the diagnostic target and the potential vaccine candidate for eliciting neutralizing antibodies and protective immune responses. In response to the escalating Mpox outbreak, three monoclonal antibodies (mAbs) (2-9B, 3-8G, and 2-5H) targeting the different domains of MPXV A29L have been developed in the study. Among them, 2-5H is highly specific for MPXV A29L without exhibiting cross-reactivity with VACV A27L. The antibody pairing composed of 2-5H and 3-8G has been developed as the lateral flow immunochromatographic assay for specific detection of MPXV A29L. However, these three mAbs were unable to inhibit A29L binding to heparin column or prevent MPXV infection in the neutralization test assays. The results of the serological assays using the truncated A29L fragments as the antigens showed that the Mpox patient sera contained significantly lower levels of antibodies targeting the N-terminal 1-34 residues of A29L, suggesting that the N-terminal portion of A29L is less immunogenic upon natural infection. KEY POINTS: • MAbs 2-9B, 3-8G, and 2-5H neither interrupted A29L binding to heparin nor neutralized MPXV. • The LFIA composed of 3-8G and 2-5H can specifically distinguish MPXV A29L from VACV A27L. • Mpox patient sera contained lower levels of antibodies targeting the N-terminal portion of A29L.
Collapse
Affiliation(s)
- Chia-Yu Liang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Tai-Ling Chao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Chong-Syun Chao
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Wang-Da Liu
- Department of Internal Medicine, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, 100, Taiwan
- Department of Medicine, National Taiwan University Cancer Center, Taipei 106, Taiwan
| | - Yu-Chen Cheng
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
- Department of Laboratory Medicine, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, 100, Taiwan.
| | - Shih-Chung Chang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan.
- Center of Biotechnology, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
5
|
Garcia-Atutxa I, Mondragon-Teran P, Huerta-Saquero A, Villanueva-Flores F. Advancements in monkeypox vaccines development: a critical review of emerging technologies. Front Immunol 2024; 15:1456060. [PMID: 39464881 PMCID: PMC11502315 DOI: 10.3389/fimmu.2024.1456060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/20/2024] [Indexed: 10/29/2024] Open
Abstract
Monkeypox (mpox) is a zoonotic illness caused by the monkeypox virus (MPXV), with higher health concerns among people who are pregnant, children, and persons who are immunocompromised, including people with untreated and advanced HIV disease. Significant progress has been made in developing vaccines against mpox, yet critical challenges and limitations persist in ensuring their effectiveness, safety, and accessibility. The pertinence of this review is highlighted by the World Health Organization's declaration of a global health emergency on August 14, 2024, due to the recent mpox outbreak, underscoring the critical necessity for effective vaccine solutions in the face of a rapidly evolving virus. Here, we comprehensively analyze various vaccine platforms utilized in mpox prevention, including attenuated and non-replicating virus vaccines, viral vector-based vaccines, recombinant protein vaccines, and DNA and mRNA vaccines. We evaluate the advantages and limitations of each platform, highlighting the urgent need for ongoing research and innovation to enhance vaccine efficacy and safety. Recent advancements, such as incorporating immunostimulatory sequences, improved delivery systems, and developing polyvalent vaccines, are explored for their potential to offer broader protection against diverse orthopoxvirus strains. This work underscores the need to optimize currently available vaccines and investigate novel vaccination strategies to address future public health emergencies effectively. By focusing on these advanced methodologies, we aim to contribute to the development of robust and adaptable vaccine solutions for mpox and other related viral threats.
Collapse
Affiliation(s)
- Igor Garcia-Atutxa
- Computer Science Department, Universidad Católica de Murcia (UCAM), Murcia, Spain
| | - Paul Mondragon-Teran
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| | - Alejandro Huerta-Saquero
- Departamento de Bionanotecnología, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México (UNAM), Ensenada, Mexico
| | - Francisca Villanueva-Flores
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| |
Collapse
|
6
|
Pattanaik A, Lodha L, Marate S, K D, Sushma Bhandarkar B, V S, Ashtaputre N, Mani RS. Buffalopox: An emerging zoonotic challenge. Infect Dis Now 2024; 54:104954. [PMID: 39033879 DOI: 10.1016/j.idnow.2024.104954] [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: 03/28/2024] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
As a variant of Vaccinia virus, Buffalopox virus is known to cause Buffalopox disease. In recent times, sporadic outbreaks of the infection in humans have been reported, especially in the endemic countries of Southeast Asia. Though mortality has not been high, associated morbidity is significant. Due to waning cross-protective immunity against smallpox, Buffalopox virus is one of the several orthopox viruses likely to emerge or reemerge. To combat this virus, early recognition, isolation, and management of the infection in animals and humans is of prime importance. In addition, vaccination in animals and humans at risk of acquiring infection is essential as a means of limiting animal-to-animal and animal-to-human spread of the virus. With this in mind, a collaborative approach between the animal and human health sectors is indispensable.
Collapse
Affiliation(s)
- Amrita Pattanaik
- Manipal Institute of Virology, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576 104, India
| | - Lonika Lodha
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka 560029, India
| | - Srilatha Marate
- Manipal Institute of Virology, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576 104, India
| | - Dhanya K
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka 560029, India
| | - B Sushma Bhandarkar
- Manipal Institute of Virology, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576 104, India
| | - Sreelakshmi V
- Manipal Institute of Virology, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576 104, India
| | - Nidhi Ashtaputre
- Manipal Institute of Virology, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576 104, India
| | - Reeta S Mani
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka 560029, India.
| |
Collapse
|
7
|
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
|
8
|
Lee MH, Choi HS, Kim NY, Sim E, Choi JY, Hong S, Shin YK, Yu CH, Gu SH, Song DH, Hur GH, Shin S. Post-Vaccination Delivery of CpG ODNs Enhances the Th2-Associated Protective Immunity of the Smallpox DNA Vaccine. Mol Biotechnol 2024; 66:1718-1726. [PMID: 37428433 DOI: 10.1007/s12033-023-00800-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 06/15/2023] [Indexed: 07/11/2023]
Abstract
Potential threat of smallpox bioterrorism and concerns related to the adverse effects of currently licensed live-virus vaccines suggest the need to develop novel vaccines with better efficacy against smallpox. Use of DNA vaccines containing specific antigen-encoding plasmids prevents the risks associated with live-virus vaccines, offering a promising alternative to conventional smallpox vaccines. In this study, we investigated the efficiency of toll-like receptor (TLR) ligands in enhancing the immunogenicity of smallpox DNA vaccines. BALB/c mice were immunized with a DNA vaccine encoding the vaccinia virus L1R protein, along with the cytosine-phosphate-guanine (CpG) motif as a vaccine adjuvant, and their immune response was analyzed. Administration of B-type CpG oligodeoxynucleotides (ODNs) as TLR9 ligands 24 h after DNA vaccination enhanced the Th2-biased L1R-specific antibody immunity in mice. Moreover, B-type CpG ODNs improved the protective effects of the DNA vaccine against the lethal Orthopoxvirus challenge. Therefore, use of L1R DNA vaccines with CpG ODNs as adjuvants is a promising approach to achieve effective immunogenicity against smallpox infection.
Collapse
Affiliation(s)
- Min Hoon Lee
- R&D Center, ABION Inc., Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | | | - Na Young Kim
- R&D Center, ABION Inc., Seoul, Republic of Korea
| | - Euni Sim
- R&D Center, ABION Inc., Seoul, Republic of Korea
| | | | - Sungyoul Hong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young Kee Shin
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Chi Ho Yu
- Chem-Bio Technology Center, Agency for Defense Development, Daejeon, Republic of Korea
| | - Se Hun Gu
- Chem-Bio Technology Center, Agency for Defense Development, Daejeon, Republic of Korea
| | - Dong Hyun Song
- Chem-Bio Technology Center, Agency for Defense Development, Daejeon, Republic of Korea
| | - Gyueng Haeng Hur
- Chem-Bio Technology Center, Agency for Defense Development, Daejeon, Republic of Korea
| | - Sungho Shin
- Bio-MAX/N-Bio, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Wang H, Yin P, Zheng T, Qin L, Li S, Han P, Qu X, Wen J, Ding H, Wu J, Kong T, Gao Z, Hu S, Zhao X, Cao X, Fang M, Qi J, Xi JJ, Duan K, Yang X, Zhang Z, Wang Q, Tan W, Gao GF. Rational design of a 'two-in-one' immunogen DAM drives potent immune response against mpox virus. Nat Immunol 2024; 25:307-315. [PMID: 38182667 DOI: 10.1038/s41590-023-01715-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 11/17/2023] [Indexed: 01/07/2024]
Abstract
The global outbreak of the mpox virus (MPXV) in 2022 highlights the urgent need for safer and more accessible new-generation vaccines. Here, we used a structure-guided multi-antigen fusion strategy to design a 'two-in-one' immunogen based on the single-chain dimeric MPXV extracellular enveloped virus antigen A35 bivalently fused with the intracellular mature virus antigen M1, called DAM. DAM preserved the natural epitope configuration of both components and showed stronger A35-specific and M1-specific antibody responses and in vivo protective efficacy against vaccinia virus (VACV) compared to co-immunization strategies. The MPXV-specific neutralizing antibodies elicited by DAM were 28 times higher than those induced by live VACV vaccine. Aluminum-adjuvanted DAM vaccines protected mice from a lethal VACV challenge with a safety profile, and pilot-scale production confirmed the high yield and purity of DAM. Thus, our study provides innovative insights and an immunogen candidate for the development of alternative vaccines against MPXV and other orthopoxviruses.
Collapse
Affiliation(s)
- Han Wang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China.
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, School of Engineering Medicine, Beihang University, Beijing, China.
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - Peng Yin
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, School of Engineering Medicine, Beihang University, Beijing, China
| | - Tingting Zheng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lanju Qin
- Department of Biological Sciences, School of life Science, Liaoning University, Shenyang, China
| | - Shihua Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Pu Han
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xiao Qu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jun Wen
- Shanghai Junshi Biosciences, Shanghai, China
| | - Haoyi Ding
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Jiahao Wu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | | | - Zhengrong Gao
- Shenzhen Children's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Songtao Hu
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xin Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xiangyu Cao
- Department of Biological Sciences, School of life Science, Liaoning University, Shenyang, China
| | - Min Fang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jianzhong Jeff Xi
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
| | - Kai Duan
- Wuhan Institute of Biological Products, Wuhan, China
| | | | | | - Qihui Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - Wenjie Tan
- National Institute for Viral Disease Control and Prevention, China CDC, Beijing, China.
| | - George Fu Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
10
|
Riccardo V, Pablo GC. Neutralization Determinants on Poxviruses. Viruses 2023; 15:2396. [PMID: 38140637 PMCID: PMC10747254 DOI: 10.3390/v15122396] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Smallpox was a highly contagious disease caused by the variola virus. The disease affected millions of people over thousands of years and variola virus ranked as one of the deadliest viruses in human history. The complete eradication of smallpox in 1980, a major triumph in medicine, was achieved through a global vaccination campaign using a less virulent poxvirus, vaccinia virus. Despite this success, the herd immunity established by this campaign has significantly waned, and concerns are rising about the potential reintroduction of variola virus as a biological weapon or the emergence of zoonotic poxviruses. These fears were further fueled in 2022 by a global outbreak of monkeypox virus (mpox), which spread to over 100 countries, thereby boosting interest in developing new vaccines using molecular approaches. However, poxviruses are complex and creating modern vaccines against them is challenging. This review focuses on the structural biology of the six major neutralization determinants on poxviruses (D8, H3, A27, L1, B5, and A33), the localization of epitopes targeted by neutralizing antibodies, and their application in the development of subunit vaccines.
Collapse
Affiliation(s)
| | - Guardado-Calvo Pablo
- Structural Biology of Infectious Diseases Unit, Institut Pasteur, Université Paris Cité, F-75015 Paris, France;
| |
Collapse
|
11
|
Li M, Ren Z, Wang Y, Jiang Y, Yang M, Li D, Chen J, Liang Z, Lin Y, Zeng Z, Xu R, Wang Y, Zhu L, Xiao W, Wu Q, Zhang B, Wan C, Yang Y, Wu B, Peng J, Zhao W, Shen C. Three neutralizing mAbs induced by MPXV A29L protein recognizing different epitopes act synergistically against orthopoxvirus. Emerg Microbes Infect 2023; 12:2223669. [PMID: 37288876 PMCID: PMC10286687 DOI: 10.1080/22221751.2023.2223669] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/06/2023] [Indexed: 06/09/2023]
Abstract
The worldwide outbreak of the monkeypox virus (MPXV) has become a "Public Health Emergency of International Concern" (PHEIC). Severe monkeypox virus infection can be fatal, however, effective therapeutic methods are yet to be developed. Mice were immunized with A35R protein and A29L protein of MPXV, and the binding and neutralizing activities of the immune sera against poxvirus-associated antigens and viruses were identified. A29L protein and A35R protein-specific monoclonal antibodies (mAbs) were generated and their antiviral activities of these mAbs were characterized in vitro and in vivo. Immunization with the MPXV A29L protein and A35R protein induced neutralizing antibodies against the orthopoxvirus in mice. None of the mAbs screened in this study against A35R could effectively neutralize the vaccinia virus (VACV), while three mAbs against A29L protein, 9F8, 3A1 and 2D1 were confirmed to have strong broad binding and neutralizing activities against orthopoxvirus, among which 9F8 showed the best neutralizing activity. 9F8, 3A1, and 2D1 recognized different epitopes on MPXV A29L protein, showing synergistic antiviral activity in vitro against the VACV Tian Tan and WR strains; the best activity was observed when the three antibodies were combined. In the vivo antiviral prophylactic and therapeutic experiments, 9F8 showed complete protective activity, whereas 3A1 and 2D1 showed partial protective activity. Similarly, the three antibodies showed synergistic antiviral protective activity against the two VACVs. In conclusion, three mAbs recognized different epitopes on MPXV A29L protein were developed and showed synergistic effects against orthopoxvirus.
Collapse
Affiliation(s)
- Mengjun Li
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Zuning Ren
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yuelin Wang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Yushan Jiang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Minghui Yang
- Advanced Research Institute of Multidisciplinary Sciences, Beijing Institute of Technology, Beijing, People’s Republic of China
| | - Delin Li
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, People’s Republic of China
| | - Jiayin Chen
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Zuxin Liang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Yuhao Lin
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Zhujun Zeng
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
- Medical Laboratory Dept, Guangdong Provincial Hospital of Chinese Medicine, Zhuhai, People’s Republic of China
| | - Rui Xu
- Medical Laboratory Dept, Guangdong Provincial Hospital of Chinese Medicine, Zhuhai, People’s Republic of China
| | - Yiting Wang
- Department of Laboratory, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, People’s Republic of China
| | - Li Zhu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Weiwei Xiao
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Qinghua Wu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Bao Zhang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Chengsong Wan
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and Immunity, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Bo Wu
- Hoyotek Biomedical Co., Ltd., Tianjin, People’s Republic of China
| | - Jie Peng
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Wei Zhao
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Chenguang Shen
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
12
|
Otter AD, Jones S, Hicks B, Bailey D, Callaby H, Houlihan C, Rampling T, Gordon NC, Selman H, Satheshkumar PS, Townsend M, Mehta R, Pond M, Jones R, Wright D, Oeser C, Tonge S, Linley E, Hemingway G, Coleman T, Millward S, Lloyd A, Damon I, Brooks T, Vipond R, Rowe C, Hallis B. Monkeypox virus-infected individuals mount comparable humoral immune responses as Smallpox-vaccinated individuals. Nat Commun 2023; 14:5948. [PMID: 37741831 PMCID: PMC10517934 DOI: 10.1038/s41467-023-41587-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 09/11/2023] [Indexed: 09/25/2023] Open
Abstract
In early 2022, a cluster of monkeypox virus (MPXV) infection (mpox) cases were identified within the UK with no prior travel history to MPXV-endemic regions. Subsequently, case numbers exceeding 80,000 were reported worldwide, primarily affecting gay, bisexual, and other men who have sex with men (GBMSM). Public health agencies worldwide have offered the IMVANEX Smallpox vaccination to these individuals at high-risk to provide protection and limit the spread of MPXV. We have developed a comprehensive array of ELISAs to study poxvirus-induced antibodies, utilising 24 MPXV and 3 Vaccinia virus (VACV) recombinant antigens. Panels of serum samples from individuals with differing Smallpox-vaccine doses and those with prior MPXV infection were tested on these assays, where we observed that one dose of Smallpox vaccination induces a low number of antibodies to a limited number of MPXV antigens but increasing with further vaccination doses. MPXV infection induced similar antibody responses to diverse poxvirus antigens observed in Smallpox-vaccinated individuals. We identify MPXV A27 as a serological marker of MPXV-infection, whilst MPXV M1 (VACV L1) is likely IMVANEX-specific. Here, we demonstrate analogous humoral antigen recognition between both MPXV-infected or Smallpox-vaccinated individuals, with binding to diverse yet core set of poxvirus antigens, providing opportunities for future vaccine (e.g., mRNA) and therapeutic (e.g., mAbs) design.
Collapse
Affiliation(s)
- Ashley D Otter
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK.
| | - Scott Jones
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Bethany Hicks
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Daniel Bailey
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Helen Callaby
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Catherine Houlihan
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
- Department of Infection and Immunity, University College London, London, UK
| | - Tommy Rampling
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
- The Hospital for Tropical Diseases, University College London Hospital, London, UK
- NIHR University College London Hospitals BRC, London, UK
| | - Nicola Claire Gordon
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Hannah Selman
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | | | - Michael Townsend
- Poxvirus and Rabies Branch, Centre for Disease Control and Prevention, Atlanta, GA, USA
| | - Ravi Mehta
- Imperial College Healthcare NHS Trust, London, UK
| | - Marcus Pond
- Imperial College Healthcare NHS Trust, London, UK
| | - Rachael Jones
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Deborah Wright
- Research and Development, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Clarissa Oeser
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, Colindale, London, UK
| | - Simon Tonge
- Seroepidemiology Unit, UK Health Security Agency, Manchester, UK
| | - Ezra Linley
- Seroepidemiology Unit, UK Health Security Agency, Manchester, UK
| | - Georgia Hemingway
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Tom Coleman
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Sebastian Millward
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Aaron Lloyd
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Inger Damon
- Poxvirus and Rabies Branch, Centre for Disease Control and Prevention, Atlanta, GA, USA
| | - Tim Brooks
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Richard Vipond
- Research and Development, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Cathy Rowe
- Emerging Pathogen Serology group, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Bassam Hallis
- Research and Development, UK Health Security Agency, Porton Down, Wiltshire, UK
| |
Collapse
|
13
|
Wang Y, Yang K, Zhou H. Immunogenic proteins and potential delivery platforms for mpox virus vaccine development: A rapid review. Int J Biol Macromol 2023; 245:125515. [PMID: 37353117 PMCID: PMC10284459 DOI: 10.1016/j.ijbiomac.2023.125515] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Since May 2022, the mpox virus (MPXV) has spread worldwide and become a potential threat to global public health. Vaccines are important tools for preventing MPXV transmission and infection in the population. However, there are still no available potent and applicable vaccines specifically for MPXV. Herein, we highlight several potential vaccine targets for MPVX and emphasize potent immunogens, such as M1R, E8L, H3L, A29L, A35R, and B6R proteins. These proteins can be integrated into diverse vaccine platforms to elicit powerful B-cell and T-cell responses, thereby providing protective immunity against MPXV infection. Overall, research on the MPXV vaccine targets would provide valuable information for developing timely effective MPXV-specific vaccines.
Collapse
Affiliation(s)
- Yang Wang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Kaiwen Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Hao Zhou
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China.
| |
Collapse
|
14
|
Gao F, He C, Liu M, Yuan P, Tian S, Zheng M, Zhang L, Zhou X, Xu F, Luo J, Li X. Cross-reactive immune responses to monkeypox virus induced by MVA vaccination in mice. Virol J 2023; 20:126. [PMID: 37337226 PMCID: PMC10278293 DOI: 10.1186/s12985-023-02085-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/28/2023] [Indexed: 06/21/2023] Open
Abstract
Mpox (monkeypox) infection cases increased recently in non-Mpox outbreak areas, potentially causing an international threat. The desire to defend against a potential outbreak has led to renewed efforts to develop Mpox vaccines. In this report, mice were immunized with various doses of modified vaccinia virus Ankara (MVA) to evaluate the cross-reactive immune response of MVA immunization against protective antigens of the current monkeypox virus. We demonstrated that MVA induced specific antibodies against protective antigens (A29, A35, B6, M1, H3, and I1), mediating the neutralization abilities against the MVA and the monkeypox virus (MPXV). Moreover, recombinant protective antigens of the MPXV elicited cross-binding and cross-neutralizing activities for MVA. Hence, the MVA induced cross-reactive immune responses, which may guide future efforts to develop vaccines against the recent MPXV. Notably, compared to the other protective antigens, the predominant A29 and M1 antigens mediated higher cross-neutralizing immune responses against the MVA, which could serve as antigen targets for novel orthologous orthopoxvirus vaccine.
Collapse
Affiliation(s)
- Feixia Gao
- Shanghai Institute of Biological Products, Shanghai, China
| | - Cheng He
- Shanghai Institute of Biological Products, Shanghai, China
| | - Min Liu
- Shanghai Institute of Biological Products, Shanghai, China
| | - Ping Yuan
- Shanghai Institute of Biological Products, Shanghai, China
| | - Shihua Tian
- Shanghai Institute of Biological Products, Shanghai, China
| | - Mei Zheng
- Shanghai Institute of Biological Products, Shanghai, China
| | - Linya Zhang
- Shanghai Institute of Biological Products, Shanghai, China
| | - Xu Zhou
- Shanghai Institute of Biological Products, Shanghai, China
| | | | - Jian Luo
- Shanghai Institute of Biological Products, Shanghai, China.
| | - Xiuling Li
- Shanghai Institute of Biological Products, Shanghai, China.
| |
Collapse
|
15
|
Feng Y, Zhang Y, Liu S, Guo M, Huang H, Guo C, Wang W, Zhang W, Tang H, Wan Y. Unexpectedly higher levels of anti-orthopoxvirus neutralizing antibodies are observed among gay men than general adult population. BMC Med 2023; 21:183. [PMID: 37189197 DOI: 10.1186/s12916-023-02872-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND The confirmed cases in the current outbreak of Monkeypox are predominantly identified in the networks of men who have sex with men (MSM). The preexisting antibodies may profoundly impact the transmission of monkeypox virus (MPXV), however the current-day prevalence of antibodies against MPXV among gay men is not well characterized. METHODS A cohort of gay men (n = 326) and a cohort of the general adult population (n = 295) were enrolled in this study. Binding antibodies responses against MPXV/vaccinia and neutralizing antibody responses against vaccinia virus (Tiantan strain) were measured. The antibody responses of these two cohorts were then compared, as well as the responses of individuals born before and in/after 1981 (when the smallpox vaccination ceased in China). Finally, the correlation between the anti-MPXV antibody responses and the anti-vaccinia antibody responses, and the associations between preexisting anti-orthopoxvirus antibody responses and the diagnosed sexually transmitted infections (STIs) in the MSM cohort were analyzed separately. RESULTS Our data showed that binding antibodies against MPXV H3, A29, A35, E8, B6, M1 proteins and vaccinia whole-virus lysate could be detected in individuals born both before and in/after 1981, of which the prevalence of anti-vaccinia binding antibodies was significantly higher among individuals born before 1981 in the general population cohort. Moreover, we unexpectedly found that the positive rates of binding antibody responses against MPXV H3, A29, A35, E8 and M1 proteins were significantly lower among individuals of the MSM cohort born in/after 1981, but the positive rates of anti-MPXV B6 and anti-vaccinia neutralizing antibody responses were significantly higher among these individuals compared to those of age-matched participants in the general population cohort. Additionally, we demonstrated that the positive and negative rates of anti-MPXV antibody responses were associated with the anti-vaccinia antibody responses among individuals born before 1981 in the general population cohort, but no significant association was observed among individuals born in/after 1981 in both cohorts. The positive rates of both the binding and the neutralizing antibody responses were comparable between individuals with and without diagnosed STIs in the MSM cohort. CONCLUSIONS Anti-MPXV and anti-vaccinia antibodies could be readily detected in an MSM cohort and a general population cohort. And a higher level of anti-vaccinia neutralizing antibody responses was observed among individuals who did not get vaccinated against smallpox in the MSM cohort compared to age-matched individuals in the general population cohort.
Collapse
Affiliation(s)
- Yanmeng Feng
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430065, China
| | - Yifan Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan Province, Zhengzhou, 450052, China
| | - Shengya Liu
- Shenzhen International Travel Health Care Center (Shenzhen Customs District Port Outpatient Clinics), Shenzhen Customs District, Shenzhen, 518033, China
| | - Meng Guo
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430065, China
| | - Haojie Huang
- Wuhan Pioneer Social Work Service Center, Wuhan, 430071, China
| | - Cuiyuan Guo
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan Province, Zhengzhou, 450052, China
| | - Wanhai Wang
- Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan Province, Zhengzhou, 450052, China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
- Shanghai Huashen Institute of Microbes and Infections, 6 Lane 1220 Huashan Rd., Shanghai, 200052, NO, China.
| | - Heng Tang
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430065, China.
| | - Yanmin Wan
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
- Shanghai Huashen Institute of Microbes and Infections, 6 Lane 1220 Huashan Rd., Shanghai, 200052, NO, China.
- Department of Radiology, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| |
Collapse
|
16
|
Zhang Y, Zhou Y, Pei R, Chen X, Wang Y. Potential threat of human pathogenic orthopoxviruses to public health and control strategies. JOURNAL OF BIOSAFETY AND BIOSECURITY 2023; 5:1-7. [PMID: 36624850 PMCID: PMC9811937 DOI: 10.1016/j.jobb.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/19/2022] [Accepted: 12/25/2022] [Indexed: 01/06/2023] Open
Abstract
Orthopoxviruses (OPXVs) belong to a group of nucleo-cytoplasmic large DNA viruses. Human pathogenic OPXVs (hpOPXVs) include at least five viruses, among which smallpox virus and monkeypox virus are the most dangerous viral pathogens. Both viruses are classified as category-one human infectious pathogens in China. Although smallpox was globally eradicated in the 1980 s, it is still a top biosecurity threat owing to the possibility of either being leaked to the outside world from a laboratory or being weaponized by terrorists. Beginning in early May 2022, a sudden outbreak of monkeypox was concurrently reported in more than 100 disparate geographical areas, representing a public health emergency of international concern, as declared by the World Health Organization (WHO). In this review, we present the reasons for hpOPXVs such as monkeypox virus presenting a potential threat to public health. We then systematically review the historical and recent development of vaccines and drugs against smallpox and monkeypox. In the final section, we highlight the importance of viromics studies as an integral part of a forward defense strategy to eliminate the potential threat to public health from emerging or re-emerging hpOPXVs and their variants.
Collapse
Affiliation(s)
- Yongli Zhang
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences. 44 Hongshancelu Avenue, Wuhan 430071, China
| | - Yuan Zhou
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences. 44 Hongshancelu Avenue, Wuhan 430071, China
| | - Rongjuan Pei
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences. 44 Hongshancelu Avenue, Wuhan 430071, China
| | - Xinwen Chen
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences. 44 Hongshancelu Avenue, Wuhan 430071, China,Innovation Center for Pathogen Research, Guangzhou Laboratory, Guangzhou 510320, China
| | - Yun Wang
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences. 44 Hongshancelu Avenue, Wuhan 430071, China,Corresponding author
| |
Collapse
|
17
|
Waqas M, Aziz S, Liò P, Khan Y, Ali A, Iqbal A, Khan F, Almajhdi FN. Immunoinformatics design of multivalent epitope vaccine against monkeypox virus and its variants using membrane-bound, enveloped, and extracellular proteins as targets. Front Immunol 2023; 14:1091941. [PMID: 36776835 PMCID: PMC9908764 DOI: 10.3389/fimmu.2023.1091941] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
Introduction The current monkeypox (MPX) outbreak, caused by the monkeypox virus (MPXV), has turned into a global concern, with over 59,000 infection cases and 23 deaths worldwide. Objectives Herein, we aimed to exploit robust immunoinformatics approach, targeting membrane-bound, enveloped, and extracellular proteins of MPXV to formulate a chimeric antigen. Such a strategy could similarly be applied for identifying immunodominant epitopes and designing multi-epitope vaccine ensembles in other pathogens responsible for chronic pathologies that are difficult to intervene against. Methods A reverse vaccinology pipeline was used to select 11 potential vaccine candidates, which were screened and mapped to predict immunodominant B-cell and T-cell epitopes. The finalized epitopes were merged with the aid of suitable linkers, an adjuvant (Resuscitation-promoting factor), a PADRE sequence (13 aa), and an HIV TAT sequence (11 aa) to formulate a multivalent epitope vaccine. Bioinformatics tools were employed to carry out codon adaptation and computational cloning. The tertiary structure of the chimeric vaccine construct was modeled via I-TASSER, and its interaction with Toll-like receptor 4 (TLR4) was evaluated using molecular docking and molecular dynamics simulation. C-ImmSim server was implemented to examine the immune response against the designed multi-epitope antigen. Results and discussion The designed chimeric vaccine construct included 21 immunodominant epitopes (six B-cell, eight cytotoxic T lymphocyte, and seven helper T-lymphocyte) and is predicted non-allergen, antigenic, soluble, with suitable physicochemical features, that can promote cross-protection among the MPXV strains. The selected epitopes indicated a wide global population coverage (93.62%). Most finalized epitopes have 70%-100% sequence similarity with the experimentally validated immune epitopes of the vaccinia virus, which can be helpful in the speedy progression of vaccine design. Lastly, molecular docking and molecular dynamics simulation computed stable and energetically favourable interaction between the putative antigen and TLR4. Conclusion Our results show that the multi-epitope vaccine might elicit cellular and humoral immune responses and could be a potential vaccine candidate against the MPXV infection. Further experimental testing of the proposed vaccine is warranted to validate its safety and efficacy profile.
Collapse
Affiliation(s)
- Muhammad Waqas
- Department of Biotechnology and Genetic Engineering, Hazara University, Mansehra, Pakistan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz, Nizwa, Oman
| | - Shahkaar Aziz
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
| | - Pietro Liò
- Department of Computer Science and Technology, University of Cambridge, Cambridge, United Kingdom
| | - Yumna Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
| | - Amjad Ali
- Department of Biotechnology and Genetic Engineering, Hazara University, Mansehra, Pakistan
| | - Aqib Iqbal
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
- Department of Biotechnology, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Faizullah Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz, Nizwa, Oman
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Fahad Nasser Almajhdi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
18
|
Huang Y, Mu L, Wang W. Monkeypox: epidemiology, pathogenesis, treatment and prevention. Signal Transduct Target Ther 2022; 7:373. [PMID: 36319633 PMCID: PMC9626568 DOI: 10.1038/s41392-022-01215-4] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/18/2022] [Accepted: 09/27/2022] [Indexed: 11/15/2022] Open
Abstract
Monkeypox is a zoonotic disease that was once endemic in west and central Africa caused by monkeypox virus. However, cases recently have been confirmed in many nonendemic countries outside of Africa. WHO declared the ongoing monkeypox outbreak to be a public health emergency of international concern on July 23, 2022, in the context of the COVID-19 pandemic. The rapidly increasing number of confirmed cases could pose a threat to the international community. Here, we review the epidemiology of monkeypox, monkeypox virus reservoirs, novel transmission patterns, mutations and mechanisms of viral infection, clinical characteristics, laboratory diagnosis and treatment measures. In addition, strategies for the prevention, such as vaccination of smallpox vaccine, is also included. Current epidemiological data indicate that high frequency of human-to-human transmission could lead to further outbreaks, especially among men who have sex with men. The development of antiviral drugs and vaccines against monkeypox virus is urgently needed, despite some therapeutic effects of currently used drugs in the clinic. We provide useful information to improve the understanding of monkeypox virus and give guidance for the government and relative agency to prevent and control the further spread of monkeypox virus.
Collapse
Affiliation(s)
- Yong Huang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Mu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
19
|
Ahmed SF, Sohail MS, Quadeer AA, McKay MR. Vaccinia-Virus-Based Vaccines Are Expected to Elicit Highly Cross-Reactive Immunity to the 2022 Monkeypox Virus. Viruses 2022; 14:1960. [PMID: 36146766 PMCID: PMC9506226 DOI: 10.3390/v14091960] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/03/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Beginning in May 2022, a novel cluster of monkeypox virus infections was detected in humans. This virus has spread rapidly to non-endemic countries, sparking global concern. Specific vaccines based on the vaccinia virus (VACV) have demonstrated high efficacy against monkeypox viruses in the past and are considered an important outbreak control measure. Viruses observed in the current outbreak carry distinct genetic variations that have the potential to affect vaccine-induced immune recognition. Here, by investigating genetic variation with respect to orthologous immunogenic vaccinia-virus proteins, we report data that anticipates immune responses induced by VACV-based vaccines, including the currently available MVA-BN and ACAM2000 vaccines, to remain highly cross-reactive against the newly observed monkeypox viruses.
Collapse
Affiliation(s)
- Syed Faraz Ahmed
- Department of Electrical and Electronic Engineering, University of Melbourne, Parkville, VIC 3010, Australia
| | - Muhammad Saqib Sohail
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Ahmed Abdul Quadeer
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Matthew R. McKay
- Department of Electrical and Electronic Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| |
Collapse
|
20
|
Frey SE, Stapleton JT, Ballas ZK, Rasmussen WL, Kaufman TM, Blevins TP, Jensen TL, Davies DH, Tary-Lehmann M, Chaplin P, Hill H, Goll JB. Human Antibody Responses Following Vaccinia Immunization Using Protein Microarrays and Correlation With Cell-Mediated Immunity and Antibody-Dependent Cellular Cytotoxicity Responses. J Infect Dis 2021; 224:1372-1382. [PMID: 33675226 PMCID: PMC8861366 DOI: 10.1093/infdis/jiab111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/05/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND There are limited data regarding immunological correlates of protection for the modified vaccinia Ankara (MVA) smallpox vaccine. METHODS A total of 523 vaccinia-naive subjects were randomized to receive 2 vaccine doses, as lyophilized MVA given subcutaneously, liquid MVA given subcutaneously (liquid-SC group), or liquid MVA given intradermally (liquid-ID group) 28 days apart. For a subset of subjects, antibody-dependent cellular cytotoxicity (ADCC), interferon-γ release enzyme-linked immunospot (ELISPOT), and protein microarray antibody-binding assays were conducted. Protein microarray responses were assessed for correlations with plaque reduction neutralization titer (PRNT), enzyme-linked immunosorbent assay, ADCC, and ELISPOT results. RESULTS MVA elicited significant microarray antibody responses to 15 of 224 antigens, mostly virion membrane proteins, at day 28 or 42, particularly WR113/D8L and WR101H3L. In the liquid-SC group, responses to 9 antigens, including WR113/D8L and WR101/H3L, correlated with PRNT results. Three were correlated in the liquid-ID group. No significant correlations were observed with ELISPOT responses. In the liquid-ID group, WR052/F13L, a membrane glycoprotein, correlated with ADCC responses. CONCLUSIONS MVA elicited antibodies to 15 vaccinia strain antigens representing virion membrane. Antibody responses to 2 proteins strongly increased and significantly correlated with increases in PRNT. Responses to these proteins are potential correlates of protection and may serve as immunogens for future vaccine development. CLINICAL TRIALS REGISTRATION NCT00914732.
Collapse
Affiliation(s)
- Sharon E Frey
- Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA
| | - Jack T Stapleton
- Department of Internal Medicine, University of Iowa and Iowa City VA Medical Center, Iowa City, Iowa, USA
| | - Zuhair K Ballas
- Department of Internal Medicine, University of Iowa and Iowa City VA Medical Center, Iowa City, Iowa, USA
| | - Wendy L Rasmussen
- Department of Internal Medicine, University of Iowa and Iowa City VA Medical Center, Iowa City, Iowa, USA
| | - Thomas M Kaufman
- Department of Internal Medicine, University of Iowa and Iowa City VA Medical Center, Iowa City, Iowa, USA
| | - Tammy P Blevins
- Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA
| | | | - D Huw Davies
- Vaccine Research & Development Center, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California, USA
| | | | | | | | | |
Collapse
|
21
|
Roth KDR, Wenzel EV, Ruschig M, Steinke S, Langreder N, Heine PA, Schneider KT, Ballmann R, Fühner V, Kuhn P, Schirrmann T, Frenzel A, Dübel S, Schubert M, Moreira GMSG, Bertoglio F, Russo G, Hust M. Developing Recombinant Antibodies by Phage Display Against Infectious Diseases and Toxins for Diagnostics and Therapy. Front Cell Infect Microbiol 2021; 11:697876. [PMID: 34307196 PMCID: PMC8294040 DOI: 10.3389/fcimb.2021.697876] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/21/2021] [Indexed: 12/30/2022] Open
Abstract
Antibodies are essential molecules for diagnosis and treatment of diseases caused by pathogens and their toxins. Antibodies were integrated in our medical repertoire against infectious diseases more than hundred years ago by using animal sera to treat tetanus and diphtheria. In these days, most developed therapeutic antibodies target cancer or autoimmune diseases. The COVID-19 pandemic was a reminder about the importance of antibodies for therapy against infectious diseases. While monoclonal antibodies could be generated by hybridoma technology since the 70ies of the former century, nowadays antibody phage display, among other display technologies, is robustly established to discover new human monoclonal antibodies. Phage display is an in vitro technology which confers the potential for generating antibodies from universal libraries against any conceivable molecule of sufficient size and omits the limitations of the immune systems. If convalescent patients or immunized/infected animals are available, it is possible to construct immune phage display libraries to select in vivo affinity-matured antibodies. A further advantage is the availability of the DNA sequence encoding the phage displayed antibody fragment, which is packaged in the phage particles. Therefore, the selected antibody fragments can be rapidly further engineered in any needed antibody format according to the requirements of the final application. In this review, we present an overview of phage display derived recombinant antibodies against bacterial, viral and eukaryotic pathogens, as well as microbial toxins, intended for diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Kristian Daniel Ralph Roth
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Esther Veronika Wenzel
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,Abcalis GmbH, Braunschweig, Germany
| | - Maximilian Ruschig
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Stephan Steinke
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Nora Langreder
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Philip Alexander Heine
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kai-Thomas Schneider
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Rico Ballmann
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Viola Fühner
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | | | | | | | - Stefan Dübel
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,Abcalis GmbH, Braunschweig, Germany.,YUMAB GmbH, Braunschweig, Germany
| | - Maren Schubert
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | | | - Federico Bertoglio
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Giulio Russo
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,Abcalis GmbH, Braunschweig, Germany
| | - Michael Hust
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,YUMAB GmbH, Braunschweig, Germany
| |
Collapse
|
22
|
Xiao Y, Zeng Y, Schante C, Joshi SB, Buchman GW, Volkin DB, Middaugh CR, Isaacs SN. Short-term and longer-term protective immune responses generated by subunit vaccination with smallpox A33, B5, L1 or A27 proteins adjuvanted with aluminum hydroxide and CpG in mice challenged with vaccinia virus. Vaccine 2020; 38:6007-6018. [PMID: 32741672 PMCID: PMC7456309 DOI: 10.1016/j.vaccine.2020.07.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 04/08/2020] [Accepted: 07/10/2020] [Indexed: 12/28/2022]
Abstract
Smallpox, a contagious and deadly disease caused by variola virus, was eradicated by a strategy that included vaccination with vaccinia virus, a live-virus vaccine. Because the threat of bioterrorism with smallpox persists and infections with zoonotic poxvirus infections like monkeypox continue, and there may be a time when an alternative vaccine platform is needed, recombinant-subunit vaccine strategies for poxviruses have been pursued. Our prior work focused on understanding the immune responses generated to vaccine-formulations containing the virus protein L1. In this work, we examine vaccine-formulations with additional key protein targets: A33 and B5 (components of the extracellular virus) and another protein on the mature virus (A27) adjuvanted with aluminum hydroxide (AH) with and without CpG- oligonucleotide. Each vaccine was formulated to allow either adsorption or non-adsorption of the protein (and CpG) to AH. Mice given a prime and single boost produced long-lasting antibody responses. A second boost (given ~5-months after the first) further increased antibody titers. Similar to our prior findings with L1 vaccine-formulations, the most protective A33 vaccine-formulations included CpG, resulted in the generation of IgG2a-antibody responses. Unlike the prior findings with L1 (where formulations that adsorbed both the protein and the CpG to AH resulted in 100% survival after challenge and minimal weight loss), the AH-adsorption status of A33 and CpG did not play as important a role, since both AH-adsorbed and non-adsorbed groups lost weight after challenge and had similar survival. Vaccination with B5-formulations gave different results. While CpG-containing formulations were the only ones that generated IgG2a-antibody responses, the vaccine-formulation that adsorbed B5 to AH (without CpG) was as equally effective in protecting mice after challenge. These results indicate that the mechanism of how antibodies against A33 and B5 protect differ. The data also show the complexity of designing optimized vaccine-formulations containing multiple adjuvants and recombinant protein-based antigens.
Collapse
Affiliation(s)
- Yuhong Xiao
- Perelman School of Medicine at the University of Pennsylvania, Department of Medicine, Division of Infectious Diseases, Philadelphia, PA 19104-6073, United States
| | - Yuhong Zeng
- University of Kansas, Macromolecular and Vaccine Stabilization Center, Department of Pharmaceutical Chemistry, 2030 Becker Drive, Lawrence, KS 66047, United States
| | - Carole Schante
- University of Kansas, Macromolecular and Vaccine Stabilization Center, Department of Pharmaceutical Chemistry, 2030 Becker Drive, Lawrence, KS 66047, United States
| | - Sangeeta B Joshi
- University of Kansas, Macromolecular and Vaccine Stabilization Center, Department of Pharmaceutical Chemistry, 2030 Becker Drive, Lawrence, KS 66047, United States
| | - George W Buchman
- Chesapeake-Perl, Inc., 8510 A Corridor Rd., Savage, MD 20763, United States
| | - David B Volkin
- University of Kansas, Macromolecular and Vaccine Stabilization Center, Department of Pharmaceutical Chemistry, 2030 Becker Drive, Lawrence, KS 66047, United States
| | - C Russell Middaugh
- University of Kansas, Macromolecular and Vaccine Stabilization Center, Department of Pharmaceutical Chemistry, 2030 Becker Drive, Lawrence, KS 66047, United States
| | - Stuart N Isaacs
- Perelman School of Medicine at the University of Pennsylvania, Department of Medicine, Division of Infectious Diseases, Philadelphia, PA 19104-6073, United States.
| |
Collapse
|
23
|
Characterization and Protective Activity of Monoclonal Antibodies Directed against Streptococcus suis Serotype 2 Capsular Polysaccharide Obtained Using a Glycoconjugate. Pathogens 2019; 8:pathogens8030139. [PMID: 31500262 PMCID: PMC6789524 DOI: 10.3390/pathogens8030139] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 02/06/2023] Open
Abstract
Streptococcus suis serotype 2 is an encapsulated bacterium and an important swine pathogen. Opsonizing antibody responses targeting capsular polysaccharides (CPSs) are protective against extracellular pathogens. To elucidate the protective activity of monoclonal antibodies (mAbs) directed against S. suis serotype 2 CPS, mice were immunized with a serotype 2 CPS-glycoconjugate and three hybridomas were isolated; of which, two were murine IgMs and the other a murine IgG1. Whereas the IgMs (mAbs 9E7 and 13C8) showed different reactivity levels with S. suis serotypes 1, 1/2, 2 and 14, the IgG1 (mAb 16H11) was shown to be serotype 2-specific. All mAbs targeted the sialylated chain of the CPSs. Using an opsonophagocytosis assay, the IgMs were opsonizing towards the S. suis serotypes to which they cross-react, while the IgG1 failed to induce bacterial elimination. In a model of mouse passive immunization followed by a lethal challenge with S. suis serotype 2, the IgG1 and IgM cross-reacting only with serotype 14 (mAb 13C8) failed to protect, while the IgM cross-reacting with serotypes 1, 1/2, and 14 (mAb 9E7) was shown to be protective by limiting bacteremia. These new mAbs show promise as new S. suis diagnostic tools, as well as potential for therapeutic applications.
Collapse
|
24
|
Yan J, Morrow MP, Chu JS, Racine T, Reed CC, Khan AS, Broderick KE, Kim JJ, Kobinger GP, Sardesai NY, Weiner DB. Broad cross-protective anti-hemagglutination responses elicited by influenza microconsensus DNA vaccine. Vaccine 2019; 36:3079-3089. [PMID: 29100705 DOI: 10.1016/j.vaccine.2017.09.086] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 07/28/2017] [Accepted: 09/28/2017] [Indexed: 10/18/2022]
Abstract
Despite the routine development and distribution of seasonal influenza vaccines, influenza remains an important pathogen contributing to significant human morbidity as well as mortality each year. The seasonal variability of influenza creates a significant issue for vaccine development of seasonal strains that can afford protection from infection or disease based on serotype matching. It is appreciated that the globular head of the HA antigen contained in the vaccines generates antibodies that result in HAI activity that are a major correlates of the protection against a particular strain. Due to seasonal genetic changes in the HA protein, however, new vaccine strains are needed to be developed continually to match the new HA antigen of that seasons virus. A distinct advantage in seasonal vaccine development would be if a small group of antigens could be developed that could span many seasons without needed to be replaced due to this genetic drift. Here we report on a synthetic microconsensus approach that relies on a small collection of 4 synthetic H1HA DNA antigens which together induce broad protective HAI immunity spanning decades of H1 influenza viruses in mice, guinea pigs and non-human primates. The protective HAI titers induced by microconsensus immunogens are fully functional in vivo as immunized ferrets were completely protected from A/Mexico/InDRE4487/2009 virus infection and morbidity associated with lethal challenge. These results are encouraging that a limited easy-to-formulate collection of invariant antigens can be developed which can span seasonal vaccine changes allowing for continued immune protection.
Collapse
Affiliation(s)
- Jian Yan
- Inovio Pharmaceuticals Inc, 660 W. Germantown Pike, Suite 110, Plymouth Meeting, PA 19462, USA
| | - Matthew P Morrow
- Inovio Pharmaceuticals Inc, 660 W. Germantown Pike, Suite 110, Plymouth Meeting, PA 19462, USA
| | - Jaemi S Chu
- The Wistar Institute of Anatomy & Biology, 3601 Spruce St, Philadelphia, PA 19104, USA
| | - Trina Racine
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Charles C Reed
- Inovio Pharmaceuticals Inc, 660 W. Germantown Pike, Suite 110, Plymouth Meeting, PA 19462, USA
| | - Amir S Khan
- Inovio Pharmaceuticals Inc, 660 W. Germantown Pike, Suite 110, Plymouth Meeting, PA 19462, USA
| | - Kate E Broderick
- Inovio Pharmaceuticals Inc, 660 W. Germantown Pike, Suite 110, Plymouth Meeting, PA 19462, USA
| | - J Joseph Kim
- Inovio Pharmaceuticals Inc, 660 W. Germantown Pike, Suite 110, Plymouth Meeting, PA 19462, USA
| | - Gary P Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Niranjan Y Sardesai
- Inovio Pharmaceuticals Inc, 660 W. Germantown Pike, Suite 110, Plymouth Meeting, PA 19462, USA
| | - David B Weiner
- The Wistar Institute of Anatomy & Biology, 3601 Spruce St, Philadelphia, PA 19104, USA.
| |
Collapse
|
25
|
Reeman S, Gates AJ, Pulford DJ, Krieg A, Ulaeto DO. Protection of Mice from Lethal Vaccinia Virus Infection by Vaccinia Virus Protein Subunits with a CpG Adjuvant. Viruses 2017; 9:v9120378. [PMID: 29232844 PMCID: PMC5744152 DOI: 10.3390/v9120378] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/03/2017] [Accepted: 12/04/2017] [Indexed: 12/23/2022] Open
Abstract
Smallpox vaccination carries a high risk of adverse events in recipients with a variety of contra-indications for live vaccines. Although alternative non-replicating vaccines have been described in the form of replication-deficient vaccine viruses, DNA vaccines, and subunit vaccines, these are less efficacious than replicating vaccines in animal models. DNA and subunit vaccines in particular have not been shown to give equivalent protection to the traditional replicating smallpox vaccine. We show here that combinations of the orthopoxvirus A27, A33, B5 and L1 proteins give differing levels of protection when administered in different combinations with different adjuvants. In particular, the combination of B5 and A27 proteins adjuvanted with CpG oligodeoxynucleotides (ODN) gives a level of protection in mice that is equivalent to the Lister traditional vaccine in a lethal vaccinia virus challenge model.
Collapse
Affiliation(s)
- Sarah Reeman
- Chemical, Biological & Radiological Division, Dstl Porton Down, Salisbury SP4 0JQ, UK.
| | - Amanda J Gates
- Chemical, Biological & Radiological Division, Dstl Porton Down, Salisbury SP4 0JQ, UK.
| | - David J Pulford
- Animal Health Laboratory, Ministry for Primary Industries, Wallaceville, Upper Hutt 5140, New Zealand.
| | - Art Krieg
- Checkmate Pharmaceuticals, One Broadway, 14th Floor, Cambridge, MA 02142, USA.
| | - David O Ulaeto
- Chemical, Biological & Radiological Division, Dstl Porton Down, Salisbury SP4 0JQ, UK.
- The Pirbright Institute, Pirbright GU24 0NF, UK.
| |
Collapse
|
26
|
Porcine Dendritic Cells as an In Vitro Model to Assess the Immunological Behaviour of Streptococcus suis Subunit Vaccine Formulations and the Polarizing Effect of Adjuvants. Pathogens 2017; 6:pathogens6010013. [PMID: 28327531 PMCID: PMC5371901 DOI: 10.3390/pathogens6010013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/10/2017] [Accepted: 03/18/2017] [Indexed: 01/03/2023] Open
Abstract
An in vitro porcine bone marrow-derived dendritic cell (DC) culture was developed as a model for evaluating immune polarization induced by adjuvants when administered with immunogens that may become vaccine candidates if appropriately formulated. The swine pathogen Streptococcus suis was chosen as a prototype to evaluate proposed S. suis vaccine candidates in combination with the adjuvants Poly I:C, Quil A ®, Alhydrogel ®, TiterMax Gold ® and Stimune ®. The toll-like receptor ligand Poly I:C and the saponin Quil A ® polarized swine DC cytokines towards a type 1 phenotype, with preferential production of IL-12 and TNF-α. The water-in-oil adjuvants TiterMax Gold ® and Stimune ® favoured a type 2 profile as suggested by a marked IL-6 release. In contrast, Alhydrogel ® induced a type 1/type 2 mixed cytokine profile. The antigen type differently modified the magnitude of the adjuvant effect, but overall polarization was preserved. This is the first comparative report on swine DC immune activation by different adjuvants. Although further swine immunization studies would be required to better characterize the induced responses, the herein proposed in vitro model is a promising approach that helps assessing behaviour of the vaccine formulation rapidly at the pre-screening stage and will certainly reduce numbers of animals used while advancing vaccinology science.
Collapse
|
27
|
Kumar A, Yogisharadhya R, Venkatesan G, Bhanuprakash V, Pandey AB, Shivachandra SB. Co-administration of recombinant major envelope proteins (rA27L and rH3L) of buffalopox virus provides enhanced immunogenicity and protective efficacy in animal models. Antiviral Res 2017; 141:174-178. [PMID: 28259752 DOI: 10.1016/j.antiviral.2017.02.017] [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: 12/16/2016] [Accepted: 02/27/2017] [Indexed: 01/09/2023]
Abstract
Buffalopox virus (BPXV) and other vaccinia-like viruses (VLVs) are causing an emerging/re-emerging zoonosis affecting buffaloes, cattle and humans in India and other countries. A27L and H3L are immuno-dominant major envelope proteins of intracellular mature virion (IMV) of orthopoxviruses (OPVs) and are highly conserved with an ability to elicit neutralizing antibodies. In the present study, two recombinant proteins namely; rA27L (21S to E110; ∼30 kDa) and rH3L(1M to I280; ∼50 kDa) of BPXV-Vij/96 produced from Escherichia coli were used in vaccine formulation. A combined recombinant subunit vaccine comprising rA27L and rH3L antigens (10 μg of each) was used for active immunization of adult mice (20μg/dose/mice) with or without adjuvant (FCA/FIA) by intramuscular route. Immune responses revealed a gradual increase in antigen specific serum IgG as well as neutralizing antibody titers measured by using indirect-ELISA and serum neutralization test (SNT) respectively, which were higher as compared to that elicited by individual antigens. Suckling mice passively administered with combined anti-A27L and anti-H3L sera showed a complete (100%) pre-exposure protection upon challenge with virulent BPXV. Conclusively, this study highlights the potential utility of rA27L and rH3L proteins as safer candidate prophylactic antigens in combined recombinant subunit vaccine for buffalopox as well as passive protective efficacy of combined sera in employing better pre-exposure protection against virulent BPXV.
Collapse
Affiliation(s)
- Amit Kumar
- Pox Virus Laboratory, Division of Virology, ICAR-Indian Veterinary Research Institute (IVRI), Regional Campus, Mukteswar, 263138, Nainital (District), Uttarakhand (UK), India
| | - Revanaiah Yogisharadhya
- Pox Virus Laboratory, Division of Virology, ICAR-Indian Veterinary Research Institute (IVRI), Regional Campus, Mukteswar, 263138, Nainital (District), Uttarakhand (UK), India
| | - Gnanavel Venkatesan
- Pox Virus Laboratory, Division of Virology, ICAR-Indian Veterinary Research Institute (IVRI), Regional Campus, Mukteswar, 263138, Nainital (District), Uttarakhand (UK), India
| | - Veerakyathappa Bhanuprakash
- Pox Virus Laboratory, Division of Virology, ICAR-Indian Veterinary Research Institute (IVRI), Regional Campus, Mukteswar, 263138, Nainital (District), Uttarakhand (UK), India.
| | - Awadh Bihari Pandey
- Pox Virus Laboratory, Division of Virology, ICAR-Indian Veterinary Research Institute (IVRI), Regional Campus, Mukteswar, 263138, Nainital (District), Uttarakhand (UK), India
| | - Sathish Bhadravati Shivachandra
- Pox Virus Laboratory, Division of Virology, ICAR-Indian Veterinary Research Institute (IVRI), Regional Campus, Mukteswar, 263138, Nainital (District), Uttarakhand (UK), India
| |
Collapse
|
28
|
Trindade GDS, Emerson GL, Sammons S, Frace M, Govil D, Fernandes Mota BE, Abrahão JS, de Assis FL, Olsen-Rasmussen M, Goldsmith CS, Li Y, Carroll D, Guimarães da Fonseca F, Kroon E, Damon IK. Serro 2 Virus Highlights the Fundamental Genomic and Biological Features of a Natural Vaccinia Virus Infecting Humans. Viruses 2016; 8:v8120328. [PMID: 27973399 PMCID: PMC5192389 DOI: 10.3390/v8120328] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/01/2016] [Accepted: 11/24/2016] [Indexed: 01/14/2023] Open
Abstract
Vaccinia virus (VACV) has been implicated in infections of dairy cattle and humans, and outbreaks have substantially impacted local economies and public health in Brazil. During a 2005 outbreak, a VACV strain designated Serro 2 virus (S2V) was collected from a 30-year old male milker. Our aim was to phenotypically and genetically characterize this VACV Brazilian isolate. S2V produced small round plaques without associated comets when grown in BSC40 cells. Furthermore, S2V was less virulent than the prototype strain VACV-Western Reserve (WR) in a murine model of intradermal infection, producing a tiny lesion with virtually no surrounding inflammation. The genome of S2V was sequenced by primer walking. The coding region spans 184,572 bp and contains 211 predicted genes. Mutations in envelope genes specifically associated with small plaque phenotypes were not found in S2V; however, other alterations in amino acid sequences within these genes were identified. In addition, some immunomodulatory genes were truncated in S2V. Phylogenetic analysis using immune regulatory-related genes, besides the hemagglutinin gene, segregated the Brazilian viruses into two clusters, grouping the S2V into Brazilian VACV group 1. S2V is the first naturally-circulating human-associated VACV, with a low passage history, to be extensively genetically and phenotypically characterized.
Collapse
Affiliation(s)
- Giliane de Souza Trindade
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, MG CEP 31270-901, Brazil.
| | - Ginny L Emerson
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Scott Sammons
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Michael Frace
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Dhwani Govil
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | | | - Jônatas Santos Abrahão
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, MG CEP 31270-901, Brazil.
| | - Felipe Lopes de Assis
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, MG CEP 31270-901, Brazil.
| | - Melissa Olsen-Rasmussen
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Cynthia S Goldsmith
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Yu Li
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Darin Carroll
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | | | - Erna Kroon
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, MG CEP 31270-901, Brazil.
| | - Inger K Damon
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| |
Collapse
|
29
|
Kuhn P, Fühner V, Unkauf T, Moreira GMSG, Frenzel A, Miethe S, Hust M. Recombinant antibodies for diagnostics and therapy against pathogens and toxins generated by phage display. Proteomics Clin Appl 2016; 10:922-948. [PMID: 27198131 PMCID: PMC7168043 DOI: 10.1002/prca.201600002] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/30/2016] [Accepted: 05/17/2016] [Indexed: 12/11/2022]
Abstract
Antibodies are valuable molecules for the diagnostic and treatment of diseases caused by pathogens and toxins. Traditionally, these antibodies are generated by hybridoma technology. An alternative to hybridoma technology is the use of antibody phage display to generate recombinant antibodies. This in vitro technology circumvents the limitations of the immune system and allows—in theory—the generation of antibodies against all conceivable molecules. Phage display technology enables obtaining human antibodies from naïve antibody gene libraries when either patients are not available or immunization is not ethically feasible. On the other hand, if patients or immunized/infected animals are available, it is common to construct immune phage display libraries to select in vivo affinity‐matured antibodies. Because the phage packaged DNA sequence encoding the antibodies is directly available, the antibodies can be smoothly engineered according to the requirements of the final application. In this review, an overview of phage display derived recombinant antibodies against bacterial, viral, and eukaryotic pathogens as well as toxins for diagnostics and therapy is given.
Collapse
Affiliation(s)
- Philipp Kuhn
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Viola Fühner
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Tobias Unkauf
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | | | - André Frenzel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany.,YUMAB GmbH, Braunschweig, Germany
| | - Sebastian Miethe
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany.
| |
Collapse
|
30
|
Chervyakova OV, Zaitsev VL, Iskakov BK, Tailakova ET, Strochkov VM, Sultankulova KT, Sandybayev NT, Stanbekova GE, Beisenov DK, Abduraimov YO, Mambetaliyev M, Sansyzbay AR, Kovalskaya NY, Nemchinov LG, Hammond RW. Recombinant Sheep Pox Virus Proteins Elicit Neutralizing Antibodies. Viruses 2016; 8:E159. [PMID: 27338444 PMCID: PMC4926179 DOI: 10.3390/v8060159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 05/24/2016] [Accepted: 05/30/2016] [Indexed: 12/24/2022] Open
Abstract
The aim of this work was to evaluate the immunogenicity and neutralizing activity of sheep pox virus (SPPV; genus Capripoxvirus, family Poxviridae) structural proteins as candidate subunit vaccines to control sheep pox disease. SPPV structural proteins were identified by sequence homology with proteins of vaccinia virus (VACV) strain Copenhagen. Four SPPV proteins (SPPV-ORF 060, SPPV-ORF 095, SPPV-ORF 117, and SPPV-ORF 122), orthologs of immunodominant L1, A4, A27, and A33 VACV proteins, respectively, were produced in Escherichia coli. Western blot analysis revealed the antigenic and immunogenic properties of SPPV-060, SPPV-095, SPPV-117 and SPPV-122 proteins when injected with adjuvant into experimental rabbits. Virus-neutralizing activity against SPPV in lamb kidney cell culture was detected for polyclonal antisera raised to SPPV-060, SPPV-117, and SPPV-122 proteins. To our knowledge, this is the first report demonstrating the virus-neutralizing activities of antisera raised to SPPV-060, SPPV-117, and SPPV-122 proteins.
Collapse
Affiliation(s)
- Olga V Chervyakova
- Research Institute for Biological Safety Problems, RK ME&S - Science Committee, Gvardeiskiy 080409, Kazakhstan.
| | - Valentin L Zaitsev
- Research Institute for Biological Safety Problems, RK ME&S - Science Committee, Gvardeiskiy 080409, Kazakhstan.
| | - Bulat K Iskakov
- M. A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, RK ME&S - Science Committee, Almaty 050012, Kazakhstan.
| | - Elmira T Tailakova
- Research Institute for Biological Safety Problems, RK ME&S - Science Committee, Gvardeiskiy 080409, Kazakhstan.
| | - Vitaliy M Strochkov
- Research Institute for Biological Safety Problems, RK ME&S - Science Committee, Gvardeiskiy 080409, Kazakhstan.
| | - Kulyaisan T Sultankulova
- Research Institute for Biological Safety Problems, RK ME&S - Science Committee, Gvardeiskiy 080409, Kazakhstan.
| | - Nurlan T Sandybayev
- Research Institute for Biological Safety Problems, RK ME&S - Science Committee, Gvardeiskiy 080409, Kazakhstan.
| | - Gulshan E Stanbekova
- M. A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, RK ME&S - Science Committee, Almaty 050012, Kazakhstan.
| | - Daniyar K Beisenov
- M. A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, RK ME&S - Science Committee, Almaty 050012, Kazakhstan.
| | - Yergali O Abduraimov
- Research Institute for Biological Safety Problems, RK ME&S - Science Committee, Gvardeiskiy 080409, Kazakhstan.
| | - Muratbay Mambetaliyev
- Research Institute for Biological Safety Problems, RK ME&S - Science Committee, Gvardeiskiy 080409, Kazakhstan.
| | - Abylay R Sansyzbay
- Research Institute for Biological Safety Problems, RK ME&S - Science Committee, Gvardeiskiy 080409, Kazakhstan.
| | - Natalia Y Kovalskaya
- United States Department of Agriculture, Agricultural Research Service, Molecular Plant Pathology Laboratory, Beltsville, MD 20705, USA.
| | - Lev G Nemchinov
- United States Department of Agriculture, Agricultural Research Service, Molecular Plant Pathology Laboratory, Beltsville, MD 20705, USA.
| | - Rosemarie W Hammond
- United States Department of Agriculture, Agricultural Research Service, Molecular Plant Pathology Laboratory, Beltsville, MD 20705, USA.
| |
Collapse
|
31
|
Abstract
Smallpox has shaped human history, from the earliest human civilizations well into the 20th century. With high mortality rates, rapid transmission, and serious long-term effects on survivors, smallpox was a much-feared disease. The eradication of smallpox represents an unprecedented medical victory for the lasting benefit of human health and prosperity. Concerns remain, however, about the development and use of the smallpox virus as a biological weapon, which necessitates the need for continued vaccine development. Smallpox vaccine development is thus a much-reviewed topic of high interest. This review focuses on the current state of smallpox vaccines and their context in biodefense efforts.
Collapse
Affiliation(s)
- Emily A Voigt
- a Mayo Vaccine Research Group , Mayo Clinic , Rochester , MN , USA
| | | | - Gregory A Poland
- a Mayo Vaccine Research Group , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
32
|
Böldicke T, Miethe S, Fühner V, Schirrmann T, Frenzel A, Hust M. Generation of Recombinant Antibodies Against Toxins and Viruses by Phage Display for Diagnostics and Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 917:55-76. [PMID: 27236552 PMCID: PMC7121732 DOI: 10.1007/978-3-319-32805-8_4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Antibody phage display is an in vitro technology to generate recombinant antibodies. In particular for pathogens like viruses or toxins, antibody phage display is an alternative to hybridoma technology, since it circumvents the limitations of the immune system. Phage display allows the generation of human antibodies from naive antibody gene libraries when either immunized patients are not available or immunization is not ethically feasible. This technology also allows the construction of immune libraries to select in vivo affinity matured antibodies if immunized patients or animals are available.In this review, we describe the generation of human and human-like antibodies from naive antibody gene libraries and antibodies from immune antibody gene libraries. Furthermore, we give an overview about phage display derived recombinant antibodies against viruses and toxins for diagnostics and therapy.
Collapse
Affiliation(s)
- Thomas Böldicke
- grid.7490.aRecombinant protein exprsn/Intrabdy unit, Helmholtz-Centre for Infection Rese, Braunschweig, Germany
| | - Sebastian Miethe
- Abteilung Biotechnologie, Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
| | - Viola Fühner
- Abteilung Biotechnologie, Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
| | - Thomas Schirrmann
- Abteilung Biotechnologie, Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany.,YUMAB GmbH, Rebenring 33, 38106, Braunschweig, Germany
| | - André Frenzel
- Abteilung Biotechnologie, Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany.,YUMAB GmbH, Rebenring 33, 38106, Braunschweig, Germany
| | - Michael Hust
- Abteilung Biotechnologie, Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany.
| |
Collapse
|
33
|
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
|
34
|
Matho MH, Schlossman A, Meng X, Benhnia MREI, Kaever T, Buller M, Doronin K, Parker S, Peters B, Crotty S, Xiang Y, Zajonc DM. Structural and Functional Characterization of Anti-A33 Antibodies Reveal a Potent Cross-Species Orthopoxviruses Neutralizer. PLoS Pathog 2015; 11:e1005148. [PMID: 26325270 PMCID: PMC4556652 DOI: 10.1371/journal.ppat.1005148] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/13/2015] [Indexed: 11/18/2022] Open
Abstract
Vaccinia virus A33 is an extracellular enveloped virus (EEV)-specific type II membrane glycoprotein that is essential for efficient EEV formation and long-range viral spread within the host. A33 is a target for neutralizing antibody responses against EEV. In this study, we produced seven murine anti-A33 monoclonal antibodies (MAbs) by immunizing mice with live VACV, followed by boosting with the soluble A33 homodimeric ectodomain. Five A33 specific MAbs were capable of neutralizing EEV in the presence of complement. All MAbs bind to conformational epitopes on A33 but not to linear peptides. To identify the epitopes, we have adetermined the crystal structures of three representative neutralizing MAbs in complex with A33. We have further determined the binding kinetics for each of the three antibodies to wild-type A33, as well as to engineered A33 that contained single alanine substitutions within the epitopes of the three crystallized antibodies. While the Fab of both MAbs A2C7 and A20G2 binds to a single A33 subunit, the Fab from MAb A27D7 binds to both A33 subunits simultaneously. A27D7 binding is resistant to single alanine substitutions within the A33 epitope. A27D7 also demonstrated high-affinity binding with recombinant A33 protein that mimics other orthopoxvirus strains in the A27D7 epitope, such as ectromelia, monkeypox, and cowpox virus, suggesting that A27D7 is a potent cross-neutralizer. Finally, we confirmed that A27D7 protects mice against a lethal challenge with ectromelia virus.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/metabolism
- Antibodies, Neutralizing/therapeutic use
- Antibody Affinity
- Antibody Specificity
- Antigen-Antibody Complex/chemistry
- Antigen-Antibody Complex/genetics
- Antigen-Antibody Complex/metabolism
- Chlorocebus aethiops
- Female
- Immunoglobulin Fab Fragments/chemistry
- Immunoglobulin Fab Fragments/genetics
- Immunoglobulin Fab Fragments/metabolism
- Membrane Glycoproteins/antagonists & inhibitors
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice, Inbred BALB C
- Models, Molecular
- Mutation
- Orthopoxvirus/immunology
- Orthopoxvirus/physiology
- Poxviridae Infections/immunology
- Poxviridae Infections/prevention & control
- Poxviridae Infections/virology
- Protein Conformation
- Recombinant Proteins/chemistry
- Recombinant Proteins/metabolism
- Recombinant Proteins/therapeutic use
- Vaccines, Synthetic/chemistry
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/metabolism
- Vaccines, Synthetic/therapeutic use
- Vero Cells
- Viral Envelope Proteins/antagonists & inhibitors
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/metabolism
- Viral Tropism
- Viral Vaccines/chemistry
- Viral Vaccines/genetics
- Viral Vaccines/metabolism
- Viral Vaccines/therapeutic use
Collapse
Affiliation(s)
- Michael H. Matho
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Andrew Schlossman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Xiangzhi Meng
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Mohammed Rafii-El-Idrissi Benhnia
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville; and Laboratory of Immunovirology, Unit 211, Biomedicine Institute of Seville (IBIS), Seville, Spain
| | - Thomas Kaever
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Mark Buller
- Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Konstantin Doronin
- Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Scott Parker
- Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Shane Crotty
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Yan Xiang
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Dirk M. Zajonc
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
35
|
Stepanova LA, Kotlyarov RY, Kovaleva AA, Potapchuk MV, Korotkov AV, Sergeeva MV, Kasianenko MA, Kuprianov VV, Ravin NV, Tsybalova LM, Skryabin KG, Kiselev OI. Protection against multiple influenza A virus strains induced by candidate recombinant vaccine based on heterologous M2e peptides linked to flagellin. PLoS One 2015; 10:e0119520. [PMID: 25799221 PMCID: PMC4370815 DOI: 10.1371/journal.pone.0119520] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 01/29/2015] [Indexed: 02/02/2023] Open
Abstract
Matrix 2 protein ectodomain (M2e) is considered a promising candidate for a broadly protective influenza vaccine. M2e-based vaccines against human influenza A provide only partial protection against avian influenza viruses because of differences in the M2e sequences. In this work, we evaluated the possibility of obtaining equal protection and immune response by using recombinant protein on the basis of flagellin as a carrier of the M2e peptides of human and avian influenza A viruses. Recombinant protein was generated by the fusion of two tandem copies of consensus M2e sequence from human influenza A and two copies of M2e from avian A/H5N1 viruses to flagellin (Flg-2M2eh2M2ek). Intranasal immunisation of Balb/c mice with recombinant protein significantly elicited anti-M2e IgG in serum, IgG and sIgA in BAL. Antibodies induced by the fusion protein Flg-2M2eh2M2ek bound efficiently to synthetic peptides corresponding to the human consensus M2e sequence as well as to the M2e sequence of A/Chicken/Kurgan/05/05 RG (H5N1) and recognised native M2e epitopes exposed on the surface of the MDCK cells infected with A/PR/8/34 (H1N1) and A/Chicken/Kurgan/05/05 RG (H5N1) to an equal degree. Immunisation led to both anti-M2e IgG1 and IgG2a response with IgG1 prevalence. We observed a significant intracellular production of IL-4, but not IFN-γ, by CD4+ T-cells in spleen of mice following immunisation with Flg-2M2eh2M2ek. Immunisation with the Flg-2M2eh2M2ek fusion protein provided similar protection from lethal challenge with human influenza A viruses (H1N1, H3N2) and avian influenza virus (H5N1). Immunised mice experienced significantly less weight loss and decreased lung viral titres compared to control mice. The data obtained show the potential for the development of an M2e-flagellin candidate influenza vaccine with broad spectrum protection against influenza A viruses of various origins.
Collapse
Affiliation(s)
- Liudmila A. Stepanova
- Department of Influenza Vaccines, Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
- * E-mail:
| | | | - Anna A. Kovaleva
- Department of Influenza Vaccines, Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Marina V. Potapchuk
- Department of Influenza Vaccines, Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Alexandr V. Korotkov
- Department of Influenza Vaccines, Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Mariia V. Sergeeva
- Department of Influenza Vaccines, Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Marina A. Kasianenko
- Department of Influenza Vaccines, Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | | | - Nikolai V. Ravin
- Centre “Bioengineering”, Russian Academy of Sciences, Moscow, Russia
- GenNanotech Ltd, St. Petersburg, Russia
| | - Liudmila M. Tsybalova
- Department of Influenza Vaccines, Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | | | - Oleg I. Kiselev
- Department of Influenza Vaccines, Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| |
Collapse
|
36
|
Kan S, Jia P, Sun L, Hu N, Li C, Lu H, Tian M, Qi Y, Jin N, Li X. Generation of an attenuated Tiantan vaccinia virus by deletion of the ribonucleotide reductase large subunit. Arch Virol 2014; 159:2223-31. [PMID: 24677065 DOI: 10.1007/s00705-014-2057-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 02/28/2014] [Indexed: 10/25/2022]
Abstract
Attenuation of the virulence of vaccinia Tiantan virus (VTT) underlies the strategy adopted for mass vaccination campaigns. This strategy provides advantages of safety and efficacy over traditional vaccines and is aimed at minimization of adverse health effects. In this study, a mutant form of the virus, MVTT was derived from VTT by deletion of the ribonucleotide reductase large subunit (R1) (TI4L). Compared to wild-type parental (VTT) and revertant (VTT-rev) viruses, virulence of the mutant MVTT was reduced by 100-fold based on body weight reduction and by 3,200-fold based on determination of the intracranial 50% lethal infectious dose. However, the immunogenicity of MVTT was equivalent to that of the parental VTT. We also demonstrated that the TI4L gene is not required for efficient replication. These data support the conclusion that MVTT can be used as a replicating virus vector or as a platform for the development of vaccines against infectious diseases and for cancer therapy.
Collapse
Affiliation(s)
- Shifu Kan
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences of PLA, Jilin, People's Republic of China,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Sánchez-Arreola PB, López-Uriarte S, Marichal-Gallardo PA, González-Vázquez JC, Pérez-Chavarría R, Soto-Vázquez P, López-Pacheco F, Ramírez-Medrano A, Rocha-Pizaña MR, Alvarez MM. A baseline process for the production, recovery, and purification of bacterial influenza vaccine candidates. Biotechnol Prog 2013; 29:896-908. [PMID: 23749362 DOI: 10.1002/btpr.1749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 04/08/2013] [Indexed: 11/11/2022]
Abstract
The current commercial system for influenza vaccine production depends on the culture of virus in embryonated eggs--a strategy that is both costly and poorly scalable. Consequently, a sudden pandemic event with a demand for millions of vaccine doses in a short time could readily overwhelm the available world production capacity. In this communication, we present a process that uses Escherichia coli for scalable production of recombinant vaccine candidates against influenza. A monomeric and a dimeric fragment of hemagglutinin of the influenza A H1N1/2009 virus were successfully expressed in a BL21 (DE3) pLysS variety of C41 E. coli. We present results from batch processes where induction is made with isopropyl thiogalactoside and from fed-batch experiments where expression is induced using lactose/glucose pulses. Concentrations in the range of 1.188-0.605 g/L of recombinant protein were observed in 2-L stirred tank bioreactors. The genetic construct included an N-terminal histidine tag sequence that facilitated recovery, purification, and proper refolding of the vaccine candidate by affinity chromatography in columns loaded with Ni(+2) . The proteins produced by this strategy selectively and specifically recognizes antibodies from patients diagnosed as positive to influenza A H1N1/2009. Overall protein recovery yields between 30.0 and 34.7% were typically observed. Based on these yields, a production of 4.6 × 10(3) doses L(-3) day(-1) is feasible.
Collapse
Affiliation(s)
- Pamela B Sánchez-Arreola
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey at Monterrey, Monterrey, N.L. México, C.P. 64849
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Shim BS, Choi JA, Song HH, Park SM, Cheon IS, Jang JE, Woo SJ, Cho CH, Song MS, Kim H, Song KJ, Lee JM, Kim SW, Song DS, Choi YK, Kim JO, Nguyen HH, Kim DW, Bahk YY, Yun CH, Song MK. Sublingual administration of bacteria-expressed influenza virus hemagglutinin 1 (HA1) induces protection against infection with 2009 pandemic H1N1 influenza virus. J Microbiol 2013; 51:130-5. [PMID: 23456722 DOI: 10.1007/s12275-013-2399-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 01/09/2013] [Indexed: 11/26/2022]
Abstract
Influenza viruses are respiratory pathogens that continue to pose a significantly high risk of morbidity and mortality of humans worldwide. Vaccination is one of the most effective strategies for minimizing damages by influenza outbreaks. In addition, rapid development and production of efficient vaccine with convenient administration is required in case of influenza pandemic. In this study, we generated recombinant influenza virus hemagglutinin protein 1 (sHA1) of 2009 pandemic influenza virus as a vaccine candidate using a well-established bacterial expression system and administered it into mice via sublingual (s.l.) route. We found that s.l. immunization with the recombinant sHA1 plus cholera toxin (CT) induced mucosal antibodies as well as systemic antibodies including neutralizing Abs and provided complete protection against infection with pandemic influenza virus A/CA/04/09 (H1N1) in mice. Indeed, the protection efficacy was comparable with that induced by intramuscular (i.m.) immunization route utilized as general administration route of influenza vaccine. These results suggest that s.l. vaccination with the recombinant non-glycosylated HA1 protein offers an alternative strategy to control influenza outbreaks including pandemics.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/genetics
- Administration, Sublingual
- Animals
- Antibodies, Neutralizing/analysis
- Antibodies, Neutralizing/blood
- Antibodies, Viral/analysis
- Antibodies, Viral/blood
- Cholera Toxin/administration & dosage
- Cholera Toxin/genetics
- Disease Models, Animal
- Enzyme-Linked Immunospot Assay
- Hemagglutination Inhibition Tests
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Immunity, Mucosal
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Leukocytes, Mononuclear/immunology
- Lung/virology
- Mice
- Mice, Inbred BALB C
- Orthomyxoviridae Infections/prevention & control
- Serum/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Load
Collapse
Affiliation(s)
- Byoung-Shik Shim
- Laboratory Science Division, International Vaccine Institute, Seoul 151-919, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
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
|
40
|
Structural and biochemical characterization of the vaccinia virus envelope protein D8 and its recognition by the antibody LA5. J Virol 2012; 86:8050-8. [PMID: 22623786 DOI: 10.1128/jvi.00836-12] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Smallpox vaccine is considered a gold standard of vaccines, as it is the only one that has led to the complete eradication of an infectious disease from the human population. B cell responses are critical for the protective immunity induced by the vaccine, yet their targeted epitopes recognized in humans remain poorly described. Here we describe the biochemical and structural characterization of one of the immunodominant vaccinia virus (VACV) antigens, D8, and its binding to the monoclonal antibody LA5, which is capable of neutralizing VACV in the presence of complement. The full-length D8 ectodomain was found to form a tetramer. We determined the crystal structure of the LA5 Fab-monomeric D8 complex at a resolution of 2.1 Å, as well as the unliganded structures of D8 and LA5-Fab at resolutions of 1.42 Å and 1.6 Å, respectively. D8 features a carbonic anhydrase (CAH) fold that has evolved to bind to the glycosaminoglycan (GAG) chondroitin sulfate (CS) on host cells. The central positively charged crevice of D8 was predicted to be the CS binding site by automated docking experiments. Furthermore, sequence alignment of various poxvirus D8 orthologs revealed that this crevice is structurally conserved. The D8 epitope is formed by 23 discontinuous residues that are spread across 80% of the D8 protein sequence. Interestingly, LA5 binds with a high-affinity lock-and-key mechanism above this crevice with an unusually large antibody-antigen interface, burying 2,434 Å(2) of protein surface.
Collapse
|
41
|
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
|
42
|
Hermanson G, Chun S, Felgner J, Tan X, Pablo J, Nakajima-Sasaki R, Molina DM, Felgner PL, Liang X, Davies DH. Measurement of antibody responses to Modified Vaccinia virus Ankara (MVA) and Dryvax(®) using proteome microarrays and development of recombinant protein ELISAs. Vaccine 2011; 30:614-25. [PMID: 22100890 DOI: 10.1016/j.vaccine.2011.11.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2011] [Revised: 10/27/2011] [Accepted: 11/06/2011] [Indexed: 01/14/2023]
Abstract
Modified Vaccinia virus Ankara (MVA) is an attenuated strain of vaccinia virus that is being considered as a safer alternative to replicating vaccinia vaccine strains such as Dryvax(®) and ACAM2000. Its excellent safety profile and large genome also make it an attractive vector for the delivery of heterologous genes from other pathogens. MVA was attenuated by prolonged passage through chick embryonic fibroblasts in vitro. In human and most mammalian cells, production of infectious progeny is aborted in the late stage of infection. Despite this, MVA provides high-level gene expression and is immunogenic in humans and other animals. A key issue for vaccine developers is the ability to be able to monitor an immune response to MVA in both vaccinia naïve and previously vaccinated individuals. To this end we have used antibody profiling by proteome microarray to compare profiles before and after MVA and Dryvax vaccination to identify candidate serodiagnostic antigens. Six antigens with diagnostic utility, comprising three membrane and three non-membrane proteins from the intracellular mature virion, were purified and evaluated in ELISAs. The membrane protein WR113/D8L provided the best sensitivity and specificity of the six antigens tested for monitoring both MVA and Dryvax vaccination, whereas the A-type inclusion protein homolog, WR148, provided the best discrimination. The ratio of responses to membrane protein WR132/A13L and core protein WR070/I1L also provided good discrimination between primary and secondary responses to Dryvax, whereas membrane protein WR101/H3L and virion assembly protein WR118/D13L together provided the best sensitivity for detecting antibody in previously vaccinated individuals. These data will aid the development novel MVA-based vaccines.
Collapse
|
43
|
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
|
44
|
Clark KM, Johnson JB, Kock ND, Mizel SB, Parks GD. Parainfluenza virus 5-based vaccine vectors expressing vaccinia virus (VACV) antigens provide long-term protection in mice from lethal intranasal VACV challenge. Virology 2011; 419:97-106. [PMID: 21885079 PMCID: PMC3177979 DOI: 10.1016/j.virol.2011.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 08/08/2011] [Accepted: 08/11/2011] [Indexed: 11/21/2022]
Abstract
To test the potential for parainfluenza virus 5 (PIV5)-based vectors to provide protection from vaccinia virus (VACV) infection, PIV5 was engineered to express secreted VACV L1R and B5R proteins, two important antigens for neutralization of intracellular mature (IMV) and extracellular enveloped (EEV) virions, respectively. Protection of mice from lethal intranasal VACV challenge required intranasal immunization with PIV5-L1R/B5R in a prime-boost protocol, and correlated with low VACV-induced pathology in the respiratory tract and anti-VACV neutralizing antibody. Mice immunized with PIV5-L1R/B5R showed some disease symptoms following VACV challenge such as loss of weight and hunching, but these symptoms were delayed and less severe than with unimmunized control mice. While immunization with PIV5 expressing B5R alone conferred at least some protection, the most effective immunization included the PIV5 vector expressing L1R alone or in combination with PIV5-B5R. PIV5-L1R/B5R vectors elicited protection from VACV challenge even when CD8+ cells were depleted, but not in the case of mice that were defective in B cell production. Mice were protected from VACV challenge out to at least 1.5 years after immunization with PIV5-L1R/B5R vectors, and showed significant levels of anti-VACV neutralizing antibodies. These results demonstrate the potential for PIV5-based vectors to provide long lasting protection against complex human respiratory pathogens such as VACV, but also highlight the need to understand mechanisms for the generation of strong immune responses against poorly immunogenic viral proteins.
Collapse
Affiliation(s)
- Kimberly M. Clark
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1064, USA
| | - John B. Johnson
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1064, USA
| | - Nancy D. Kock
- Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1064, USA
| | - Steven B. Mizel
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1064, USA
| | - Griffith D. Parks
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1064, USA
- Corresponding author at: Department of Microbiology and Immunology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157-1064, USA. Fax: +1 336 716 9928.
| |
Collapse
|
45
|
Huemer HP, Lassnig C, Bernhard D, Sturm S, Nowotny N, Kitchen M, Pavlic M. Cannabinoids lead to enhanced virulence of the smallpox vaccine (vaccinia) virus. Immunobiology 2011; 216:670-7. [DOI: 10.1016/j.imbio.2010.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Revised: 11/07/2010] [Accepted: 11/08/2010] [Indexed: 11/28/2022]
|
46
|
Huemer HP, Lassnig C, Nowotny N. Cowpox virus isolate virulent in humans shows attenuated phenotype in mice. Res Vet Sci 2011; 92:333-7. [PMID: 21463881 DOI: 10.1016/j.rvsc.2011.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 03/07/2011] [Accepted: 03/11/2011] [Indexed: 12/01/2022]
Abstract
We have cultured Cowpox virus (CPXV) from skin lesion material of a human patient from Austria. Phylogenetic comparison of the HA-gene revealed a rather homogeneous cluster with other local isolates from recent years, the A36R-gene was mostly related to elephant derived strains from Germany. Despite causing disease in human, the isolate AT/Carinthia/788/07 surprisingly even at high titers showed a highly reduced virulence in BALB/c mice upon intranasal inoculation as compared to vaccinia virus. This contrasts earlier reports on other CPXV isolates. Using shotgun DNA sequencing several insertions and deletions were found in genes presumably involved in host range, immune regulation as well as established virulence factors. These preliminary data could be an indication that CPXV strains with proven pathogenicity for humans may have reduced virulence in mice and vice versa. Additionally strains with a reduced virulence may have an advantage in persisting in less dense rodent populations.
Collapse
Affiliation(s)
- Hartwig P Huemer
- Department of Hygiene, Microbiology and Social Medicine, Innsbruck Medical University, Fritz-Pregl-Str. 3, R.301, A-6020 Innsbruck, Austria.
| | | | | |
Collapse
|
47
|
Monoclonal Antibody Specific to Goose Parvovirus VP3 Protein. Hybridoma (Larchmt) 2011. [DOI: 10.1089/hyb.2010.0099.mab] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
48
|
Hirao LA, Draghia-Akli R, Prigge JT, Yang M, Satishchandran A, Wu L, Hammarlund E, Khan AS, Babas T, Rhodes L, Silvera P, Slifka M, Sardesai NY, Weiner DB. Multivalent smallpox DNA vaccine delivered by intradermal electroporation drives protective immunity in nonhuman primates against lethal monkeypox challenge. J Infect Dis 2011; 203:95-102. [PMID: 21148501 PMCID: PMC3086429 DOI: 10.1093/infdis/jiq017] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 07/22/2010] [Indexed: 01/01/2023] Open
Abstract
The threat of a smallpox-based bioterrorist event or a human monkeypox outbreak has heightened the importance of new, safe vaccine approaches for these pathogens to complement older poxviral vaccine platforms. As poxviruses are large, complex viruses, they present technological challenges for simple recombinant vaccine development where a multicomponent mixtures of vaccine antigens are likely important in protection. We report that a synthetic, multivalent, highly concentrated, DNA vaccine delivered by a minimally invasive, novel skin electroporation microarray can drive polyvalent immunity in macaques, and offers protection from a highly pathogenic monkeypox challenge. Such a diverse, high-titer antibody response produced against 8 different DNA-encoded antigens delivered simultaneously in microvolumes has not been previously described. These studies represent a significant improvement in the efficiency of the DNA vaccine platform, resulting in immune responses that mimic live viral infections, and would likely have relevance for vaccine design against complex human and animal pathogens.
Collapse
Affiliation(s)
- Lauren A. Hirao
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | | | | | - Maria Yang
- Inovio Pharmaceuticals, Blue Bell, Pennsylvania
| | - Abhishek Satishchandran
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Ling Wu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Erika Hammarlund
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon
| | | | - Tahar Babas
- Southern Research Institute, 431 Aviation Way, Frederick, Maryland
| | - Lowrey Rhodes
- Southern Research Institute, 431 Aviation Way, Frederick, Maryland
| | - Peter Silvera
- Southern Research Institute, 431 Aviation Way, Frederick, Maryland
| | - Mark Slifka
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon
| | | | - David B. Weiner
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
49
|
Abstract
The eradication of smallpox, one of the great triumphs of medicine, was accomplished through the prophylactic administration of live vaccinia virus, a comparatively benign relative of variola virus, the causative agent of smallpox. Nevertheless, recent fears that variola virus may be used as a biological weapon together with the present susceptibility of unimmunized populations have spurred the development of new-generation vaccines that are safer than the original and can be produced by modern methods. Predicting the efficacy of such vaccines in the absence of human smallpox, however, depends on understanding the correlates of protection. This review outlines the biology of poxviruses with particular relevance to vaccine development, describes protein targets of humoral and cellular immunity, compares animal models of orthopoxvirus disease with human smallpox, and considers the status of second- and third-generation smallpox vaccines.
Collapse
Affiliation(s)
- Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-3210, USA.
| |
Collapse
|
50
|
Berhanu A, King DS, Mosier S, Jordan R, Jones KF, Hruby DE, Grosenbach DW. Impact of ST-246® on ACAM2000™ smallpox vaccine reactogenicity, immunogenicity, and protective efficacy in immunodeficient mice. Vaccine 2010; 29:289-303. [PMID: 21036130 PMCID: PMC3023305 DOI: 10.1016/j.vaccine.2010.10.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 09/29/2010] [Accepted: 10/13/2010] [Indexed: 11/16/2022]
Abstract
Although a highly effective vaccine against smallpox, vaccinia virus (VV) is not without adverse events, some of which can be life-threatening, particularly in immunocompromised individuals. We have recently demonstrated that the immunogenicity and protective efficacy of Dryvax(®) in immunocompetent mice is preserved even when co-administered with ST-246, an orally bioavailable small-molecule inhibitor of orthopoxvirus egress and dissemination. In addition, ST-246 markedly reduced the reactogenicity of the smallpox vaccine ACAM2000 and the highly neurovirulent VV strain Western Reserve (VV-WR). Here, we evaluated the impact of ST-246 co-administration on ACAM2000 reactogenicity, immunogenicity, and protective efficacy in seven murine models of varying degrees of humoral and cellular immunodeficiency: BALB/c and B-cell deficient (JH-KO) mice depleted of CD4(+) or CD8(+) or both subsets of T cells. We observed that ST-246 reduced vaccine lesion severity and time to complete resolution in all of the immunodeficient models examined, except in those lacking both CD4(+) and CD8(+) T cells. Although VV-specific humoral responses were moderately reduced by ST-246 treatment, cellular responses were generally comparable or slightly enhanced at both 1 and 6 months post-vaccination. Most importantly, in those models in which vaccination given alone conferred protection against lethal VV challenge, similar levels of protection were observed at both time points when vaccination was given with ST-246. These data suggest that, with the exception of individuals with irreversible, combined CD4(+) and CD8(+) T-cell deficiency, ST-246 co-administered at the time of vaccination may help reduce vaccine reactogenicity--even in those lacking humoral immunity--without impeding the induction of protective immunity.
Collapse
Affiliation(s)
- Aklile Berhanu
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | - David S. King
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | - Stacie Mosier
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | - Robert Jordan
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | - Kevin F. Jones
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | - Dennis E. Hruby
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230 Corvallis, OR 97333, USA
| | | |
Collapse
|