1
|
Crandell J, Monteiro VS, Pischel L, Fang Z, Conde L, Zhong Y, Lawres L, de Asis GM, Maciel G, Zaleski A, Lira GS, Higa LM, Breban MI, Vogels CBF, Caria J, Pinto AR, Almeida V, Maltez F, Cordeiro R, Póvoas D, Grubaugh ND, Aoun-Barakat L, Grifoni A, Sette A, Castineiras TM, Chen S, Yildirim I, Vale AM, Omer SB, Lucas C. The impact of orthopoxvirus vaccination and Mpox infection on cross-protective immunity: a multicohort observational study. THE LANCET. MICROBE 2025:101098. [PMID: 40311645 DOI: 10.1016/j.lanmic.2025.101098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/07/2025] [Accepted: 02/03/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Cross-reactive immune memory responses to orthopoxviruses in humans remain poorly characterised despite their relevance for vaccine design and outbreak control. We aimed to assess the magnitude, specificity, and durability of cross-reactive immune responses elicited by smallpox vaccines and mpox virus infection. METHODS We did a multicohort observational study involving participants from the USA, Brazil, and Portugal across four groups: Dryvax (first-generation smallpox vaccine) recipients vaccinated 40-80 years ago, JYNNEOS (third-generation smallpox vaccine) recipients vaccinated within the past year, a cohort receiving both vaccines, and patients infected with clade IIb mpox. Samples were analysed for systemic and mucosal humoral responses, neutralising antibody titres, viral antigen structural analysis, and T-cell cross-reactivity to vaccina virus, cowpox virus, and mpox virus. Statistical analyses included correlation assessments and comparisons across cohorts to determine the magnitude, longevity, and breadth of immune responses. FINDINGS Between July 7, 2022, and Aug 3, 2023, 262 participants were recruited, resulting in analysis of 378 samples. Both first-generation and third-generation smallpox vaccines elicited vaccinia virus-reactive and mpox virus-reactive antibodies, with the strongest responses targeting the less conserved extracellular virion antigens B5 and A33. Despite high concentrations of anti-mpox virus antibodies in the plasma, cross-neutralisation activity correlated with viral antigenic distance. Higher neutralisation was observed for cowpox virus than for mpox virus, which has lower antigenic conservation with vaccina virus. Complement-mediated neutralisation enhanced mpox virus neutralisation, overcoming the limitations of antigenic distance. Dryvax recipients sustained vaccina virus neutralisation titres for over 80 years, whereas cross-reactive responses did not show this durability. JYNNEOS-induced responses waned within a year. T-cell cross-reactivity was long-lasting, detected up to 70 years after vaccination. Booster vaccinations augmented the magnitude, breadth, and longevity of cross-neutralising responses. INTERPRETATION Our findings highlight the potential combined role of antibody effector functions and T-cell memory in cross-protection against orthopoxviruses. Complement-mediated neutralisation enhances cross-protection, overcoming antigenic distance. These Fc-mediated functions, along with T-cell responses, contribute to effective and long-lasting immunity conferred by smallpox vaccines against other orthopoxviruses. FUNDING Yale University and Stavros Niarchos Foundation Institute for Global Infectious Disease.
Collapse
Affiliation(s)
- Jameson Crandell
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Valter Silva Monteiro
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Lauren Pischel
- Section of Infectious Diseases, Department of Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Zhenhao Fang
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA; System Biology Institute, Yale University, West Haven, CT, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT, USA
| | - Luciana Conde
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Yi Zhong
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Lauren Lawres
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Gustavo Meira de Asis
- Laboratorio de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriela Maciel
- Laboratorio de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Agnieszka Zaleski
- Yale Center for Clinical Investigation, Yale University, New Haven, CT, USA
| | - Guilherme S Lira
- Departamento de Doenças Infecciosas e Parasitarias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiza M Higa
- Departamento de Doenças Infecciosas e Parasitarias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mallery I Breban
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Chantal B F Vogels
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Joao Caria
- Infectious Diseases Unit, Hospital de Curry Cabral, Unidade Local de Saúde São José, Lisbon, Portugal
| | - Ana Raquel Pinto
- Infectious Diseases Unit, Hospital de Curry Cabral, Unidade Local de Saúde São José, Lisbon, Portugal
| | - Vasco Almeida
- Infectious Diseases Unit, Hospital de Curry Cabral, Unidade Local de Saúde São José, Lisbon, Portugal
| | - Fernando Maltez
- Infectious Diseases Unit, Hospital de Curry Cabral, Unidade Local de Saúde São José, Lisbon, Portugal; Instituto de Saúde Ambiental-Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Rita Cordeiro
- Infectious Diseases Department, National Institute of Health Dr Ricardo Jorge, Lisbon, Portugal
| | - Diana Póvoas
- Infectious Diseases Unit, Hospital de Curry Cabral, Unidade Local de Saúde São José, Lisbon, Portugal; Lymphocyte Physiology, Instituto Gulbenkian de Ciência, Lisbon, Portugal
| | - Nathan D Grubaugh
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Lydia Aoun-Barakat
- Section of Infectious Diseases, Department of Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA; Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Terezinha M Castineiras
- Departamento de Doenças Infecciosas e Parasitarias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sidi Chen
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA; Comprehensive Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA; Stem Cell Center, Yale School of Medicine, Yale University, New Haven, CT, USA; Center for Biomedical Data Science, Yale School of Medicine, Yale University, New Haven, CT, USA; System Biology Institute, Yale University, West Haven, CT, USA; Center for Cancer Systems Biology, Yale University, West Haven, CT, USA; Wu-Tsai Institute, Yale University, New Haven, CT, USA
| | - Inci Yildirim
- Yale Institute for Global Health, Yale University, New Haven, CT, USA; Center for Infection and Immunity, Yale University, New Haven, CT, USA; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA; Department of Pediatrics, Section of Infectious Diseases and Global Health, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Andre M Vale
- Laboratorio de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Saad B Omer
- Peter O'Donnell Jr School of Public Health, University of Texas Southwestern, Dallas, TX, USA
| | - Carolina Lucas
- Department of Immunobiology, Yale School of Medicine, Yale University, New Haven, CT, USA; Center for Infection and Immunity, Yale University, New Haven, CT, USA.
| |
Collapse
|
2
|
Zhang X, He J, Shao Y. Research progress and development potential of oncolytic vaccinia virus. Chin Med J (Engl) 2025; 138:777-791. [PMID: 40097373 PMCID: PMC11970828 DOI: 10.1097/cm9.0000000000003585] [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: 10/30/2024] [Indexed: 03/19/2025] Open
Abstract
ABSTRACT Oncolytic virotherapy is a promising therapeutic approach treating tumors, where oncolytic viruses (OVs) can selectively infect and lyse tumor cells through replication, while also triggering long-lasting anti-tumor immune responses. Vaccinia virus (VV) has emerged as a leading candidate for use as an OV due to its broad cytophilicity and robust capacity to express exogenous genes. Consequently, oncolytic vaccinia virus (OVV) has entered clinical trials. This review provides an overview of the key strategies used in the development of OVV, summarizes the findings from clinical trials, and addresses the challenges that must be overcome in the advancement of OVV-based therapies. Furthermore, it explores potential future strategies for enhancing the development and clinical application of OVV, intending to improve tumor treatment outcomes. The review aims to facilitate the further development and clinical adoption of OVV, thereby advancing tumor therapies.
Collapse
Affiliation(s)
- Xinyu Zhang
- Changping Laboratory, Beijing 102206, China
- College of Life Science, Beijing Normal University, Beijing 100875, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Jiangshan He
- Changping Laboratory, Beijing 102206, China
- College of Life Science, Beijing Normal University, Beijing 100875, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Yiming Shao
- Changping Laboratory, Beijing 102206, China
- College of Life Science, Beijing Normal University, Beijing 100875, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| |
Collapse
|
3
|
Huang IH, Lai GC, Chao TL, Liu WD, Chang SY, Chang SC. Monkeypox virus H3L protein as the target antigen for developing neutralizing antibody and serological assay. Appl Microbiol Biotechnol 2025; 109:80. [PMID: 40172630 PMCID: PMC11965195 DOI: 10.1007/s00253-025-13466-6] [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: 01/22/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025]
Abstract
The large number of atypical monkeypox (Mpox) cases caused by emerging monkeypox virus (MPXV) strains was recently found in countries and regions where the Mpox was not reported before. Diagnostic tools and therapeutic agents are important countermeasures for preventing Mpox outbreak. H3L protein is the important surface antigen of MPXV for binding to host cell receptors and mediating viral infection. A broad range of murine anti-MPXV H3L monoclonal antibodies (mAbs) recognizing various binding epitopes have been generated in the study. The rapid test composed of the mAbs 4-2A and 3-3F can specifically detect H3L protein and MPXV virion. The mAb 3-3F exhibited strong MPXV neutralizing activity in a complement-dependent manner. Notably, 3-3F binds to a unique epitope within residues 35-89 of H3L protein. The serum samples collected from Mpox patients barely bound to the N-terminal portion of H3L protein ranging from 2 to 89 residues, indicating that the content of the 3-3F-like antibody is very low in Mpox patient sera. In contrast, the seropositivity was mostly observed using the C-terminal portion of H3L protein ranging from 185 to 282 residues as the target antigen in the immunoblot analysis. Taken together, the anti-MPXV H3L mAb can be developed as the Mpox diagnostic and therapeutic agents. Furthermore, H3L protein is the promising biomarker for serological analysis. KEY POINTS: •Anti-H3L mAbs can cross-react with H3L proteins in MPXV and VACV virions. •The LFIA rapid test using the mAbs 4-2A and 3-3F can specifically detect MPXV. •MPXV was neutralized by mAb 3-3F in a complement-dependent manner.
Collapse
MESH Headings
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/isolation & purification
- Animals
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Humans
- Antigens, Viral/immunology
- Monkeypox virus/immunology
- Mice
- Mpox, Monkeypox/diagnosis
- Mpox, Monkeypox/immunology
- Serologic Tests/methods
- Mice, Inbred BALB C
- Epitopes/immunology
Collapse
Affiliation(s)
- I-Hsiang Huang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Guan-Chun Lai
- 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
| | - Wang-Da Liu
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
- Department of Medicine, National Taiwan University Cancer Center, Taipei , 106, 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, National Taiwan University Hospital, College of Medicine, 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
|
4
|
Ren Z, Li M, Chen J, Gong X, Song S, Li D, Yang M, Yu J, Asghar S, Cui Y, Niu S, Liao Z, Jiang Y, Liu J, Li Y, Zhang B, Zhao W, Peng J, Yang Y, Shen C. Identification of mpox M1R and B6R monoclonal and bispecific antibodies that efficiently neutralize authentic mpox virus. Emerg Microbes Infect 2024; 13:2401931. [PMID: 39233480 DOI: 10.1080/22221751.2024.2401931] [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: 04/29/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
In 2022, the monkeypox virus (mpox virus, MPXV) exhibited global dissemination across six continents, representing a notable challenge owing to the scarcity of targeted antiviral interventions. Passive immunotherapy, such as the use of monoclonal antibodies (mAbs) and bispecific antibodies (bsAbs), has emerged as a promising option for antiviral regimens. Here, we generated several mAbs against M1R and B6R of MPXV, and subsequently characterized the antiviral activity of these antibodies both in vitro and in vivo. Two neutralizing mAbs, M1H11 and M3B2, targeting M1R, and one B6R-specific mAb, B7C9, were identified. They exhibited varying antiviral efficacy against vaccinia virus (VACV) in vitro and in vivo. A cocktail comprising M1H11 and M3B2 demonstrated a superior protective effect in vivo. A bsAb, Bis-M1M3, was engineered by conjugating the fragment crystallizable (Fc) region of the human-mouse chimeric engineered M1H11 with the single-chain fragment variable (scFv) of M3B2. In mice challenged with MPXV, Bis-M1M3 showed a notable protective effects. Analysis of neutralization mechanism showed that these mAbs and Bis-M1M3 exerted virus-neutralizing effects before the virus infects cells. In vivo pharmacokinetic experiments showed that Bis-M1M3 has a long half-life in rhesus macaques. This study provides crucial insights for further research on broad-spectrum antiviral drugs against MPXV and other orthopoxviruses.
Collapse
Affiliation(s)
- Zuning Ren
- State Key Laboratory of Organ Failure Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
- 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
| | - 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
| | - 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
| | - Xiaohua Gong
- 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
| | - Shuo Song
- 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
| | - Delin Li
- Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, People's Republic of China
| | - Minghui Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People's Republic of China
| | - Jianhai Yu
- 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
| | - Sadia Asghar
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Yanxin Cui
- 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
| | - Shiyu Niu
- 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
| | - Zhonghui Liao
- 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
| | - 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
| | - Jiahui Liu
- 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
| | - Yuqing 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
| | - 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
| | - 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
| | - Jie Peng
- State Key Laboratory of Organ Failure Research, Department of Infectious Diseases, Nanfang 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
| | - 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
- Key Laboratory of Infectious Diseases Research in South China, Southern Medical University, Ministry of Education, People's Republic of China
| |
Collapse
|
5
|
Orosco FL. Host immune responses against African swine fever virus: Insights and challenges for vaccine development. Open Vet J 2023; 13:1517-1535. [PMID: 38292721 PMCID: PMC10824091 DOI: 10.5455/ovj.2023.v13.i12.2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/22/2023] [Indexed: 02/01/2024] Open
Abstract
The African swine fever virus (ASFV) poses a serious threat to global swine populations, underscoring the urgent need for effective preventive strategies. This comprehensive review investigates the intricate interplay between innate, cellular, and humoral immunity against ASFV, with a focus on their relevance to vaccine development. By delving into immunopathogenesis and immunological challenges, this review article aims to provide a holistic perspective on the complexities of ASFV infections and immune evasion. Key findings underscore the critical role of innate immune recognition in shaping subsequent adaptive immune defenses, potential protective antigens, and the multifaceted nature of ASFV-specific antibodies and cytotoxic T-cell responses. Despite advancements, the unique attributes of ASFV present hurdles in the development of a successful vaccine. In conclusion, this review examines the current state of ASFV immune responses and offers insights into future research directions, fostering the development of effective interventions against this devastating pathogen.
Collapse
Affiliation(s)
- Fredmoore L. Orosco
- Virology and Vaccine Institute of the Philippines Program, Department of Science and Technology, Industrial Technology Development Institute, Taguig, Philippines
- S&T Fellows Program, Department of Science and Technology, Taguig, Philippines
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Manila, Philippines
| |
Collapse
|
6
|
Li M, Zhang M, Ye Q, Liu Y, Qian W. Preclinical and clinical trials of oncolytic vaccinia virus in cancer immunotherapy: a comprehensive review. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0202. [PMID: 37615308 PMCID: PMC10546091 DOI: 10.20892/j.issn.2095-3941.2023.0202] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/19/2023] [Indexed: 08/25/2023] Open
Abstract
Oncolytic virotherapy has emerged as a promising treatment for human cancers owing to an ability to elicit curative effects via systemic administration. Tumor cells often create an unfavorable immunosuppressive microenvironment that degrade viral structures and impede viral replication; however, recent studies have established that viruses altered via genetic modifications can serve as effective oncolytic agents to combat hostile tumor environments. Specifically, oncolytic vaccinia virus (OVV) has gained popularity owing to its safety, potential for systemic delivery, and large gene insertion capacity. This review highlights current research on the use of engineered mutated viruses and gene-armed OVVs to reverse the tumor microenvironment and enhance antitumor activity in vitro and in vivo, and provides an overview of ongoing clinical trials and combination therapies. In addition, we discuss the potential benefits and drawbacks of OVV as a cancer therapy, and explore different perspectives in this field.
Collapse
Affiliation(s)
- Mengyuan Li
- Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Minghuan Zhang
- Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Qian Ye
- Hangzhou Rong-Gu Biotechnology Limited Company, Hangzhou 310056, China
| | - Yunhua Liu
- Department of Pathology & Pathophysiology and Department of Surgical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wenbin Qian
- Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
7
|
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: 10] [Impact Index Per Article: 5.0] [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
|
8
|
Patel P, Nandi A, Verma SK, Kaushik N, Suar M, Choi EH, Kaushik NK. Zebrafish-based platform for emerging bio-contaminants and virus inactivation research. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162197. [PMID: 36781138 PMCID: PMC9922160 DOI: 10.1016/j.scitotenv.2023.162197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 05/27/2023]
Abstract
Emerging bio-contaminants such as viruses have affected health and environment settings of every country. Viruses are the minuscule entities resulting in severe contagious diseases like SARS, MERS, Ebola, and avian influenza. Recent epidemic like the SARS-CoV-2, the virus has undergone mutations strengthen them and allowing to escape from the remedies. Comprehensive knowledge of viruses is essential for the development of targeted therapeutic and vaccination treatments. Animal models mimicking human biology like non-human primates, rats, mice, and rabbits offer competitive advantage to assess risk of viral infections, chemical toxins, nanoparticles, and microbes. However, their economic maintenance has always been an issue. Furthermore, the redundancy of experimental results due to aforementioned aspects is also in examine. Hence, exploration for the alternative animal models is crucial for risk assessments. The current review examines zebrafish traits and explores the possibilities to monitor emerging bio-contaminants. Additionally, a comprehensive picture of the bio contaminant and virus particle invasion and abatement mechanisms in zebrafish and human cells is presented. Moreover, a zebrafish model to investigate the emerging viruses such as coronaviridae and poxviridae has been suggested.
Collapse
Affiliation(s)
- Paritosh Patel
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India; Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, 18323 Hwaseong, Republic of Korea
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| |
Collapse
|
9
|
Rabaan AA, Abas AH, Tallei TE, Al-Zaher MA, Al-Sheef NM, Fatimawali, Al-Nass EZ, Al-Ebrahim EA, Effendi Y, Idroes R, Alhabib MF, Al-Fheid HA, Adam AA, Bin Emran T. Monkeypox outbreak 2022: What we know so far and its potential drug targets and management strategies. J Med Virol 2023; 95:e28306. [PMID: 36372558 DOI: 10.1002/jmv.28306] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/28/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2022]
Abstract
Monkeypox is a rare zoonotic disease caused by infection with the monkeypox virus. The disease can result in flu-like symptoms, fever, and a persistent rash. The disease is currently spreading throughout the world and prevention and treatment efforts are being intensified. Although there is no treatment that has been specifically approved for monkeypox virus infection, infected patients may benefit from using certain antiviral medications that are typically prescribed for the treatment of smallpox. The drugs are tecovirimat, brincidofovir, and cidofovir, all of which are currently in short supply due to the spread of the monkeypox virus. Resistance is also a concern, as widespread replication of the monkeypox virus can lead to mutations that produce monkeypox viruses that are resistant to the currently available treatments. This article discusses monkeypox disease, potential drug targets, and management strategies to overcome monkeypox disease. With the discovery of new drugs, it is hoped that the problem of insufficient drugs will be resolved, and it is not anticipated that drug resistance will become a major issue in the near future.
Collapse
Affiliation(s)
- Ali A Rabaan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| | - Abdul Hawil Abas
- Faculty of Bioscience and Engineering, Ghent University, Ghent, Belgium
| | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado, North Sulawesi, Indonesia
| | - Mona A Al-Zaher
- Department of Commitment management, Directorate of Health Affairs in the Eastern Province, Dammam, Saudi Arabia
| | - Noor M Al-Sheef
- Department of Commitment management, Directorate of Health Affairs in the Eastern Province, Dammam, Saudi Arabia
| | - Fatimawali
- Pharmacy Study Program, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado, North Sulawesi, Indonesia
| | - Esraa Z Al-Nass
- Department of Commitment management, Directorate of Health Affairs in the Eastern Province, Dammam, Saudi Arabia
| | - Eba A Al-Ebrahim
- Department of Commitment management, Directorate of Health Affairs in the Eastern Province, Dammam, Saudi Arabia
| | - Yunus Effendi
- Department of Biology, Faculty of Science and Technology, Al-Azhar Indonesia University, Jakarta, Indonesia
| | - Rinaldi Idroes
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Mather F Alhabib
- Molecular Diagnostic Laboratory, Dammam Regional Laboratory and Blood Bank, Dammam, Saudi Arabia
| | - Hussain A Al-Fheid
- Molecular Diagnostic Laboratory, Dammam Regional Laboratory and Blood Bank, Dammam, Saudi Arabia
| | - Ahmad Akroman Adam
- Dentistry Study Program, Faculty of Medicine, Sam Ratulangi University, Manado, North Sulawesi, Indonesia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| |
Collapse
|
10
|
African Swine Fever Vaccinology: The Biological Challenges from Immunological Perspectives. Viruses 2022; 14:v14092021. [PMID: 36146827 PMCID: PMC9505361 DOI: 10.3390/v14092021] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/22/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
African swine fever virus (ASFV), a nucleocytoplasmic large DNA virus (NCLDV), causes African swine fever (ASF), an acute hemorrhagic disease with mortality rates up to 100% in domestic pigs. ASF is currently epidemic or endemic in many countries and threatening the global swine industry. Extensive ASF vaccine research has been conducted since the 1920s. Like inactivated viruses of other NCLDVs, such as vaccinia virus, inactivated ASFV vaccine candidates did not induce protective immunity. However, inactivated lumpy skin disease virus (poxvirus) vaccines are protective in cattle. Unlike some experimental poxvirus subunit vaccines that induced protection, ASF subunit vaccine candidates implemented with various platforms containing several ASFV structural genes or proteins failed to protect pigs effectively. Only some live attenuated viruses (LAVs) are able to protect pigs with high degrees of efficacy. There are currently several LAV ASF vaccine candidates. Only one commercial LAV vaccine is approved for use in Vietnam. LAVs, as ASF vaccines, have not yet been widely tested. Reports thus far show that the onset and duration of protection induced by the LAVs are late and short, respectively, compared to LAV vaccines for other diseases. In this review, the biological challenges in the development of ASF vaccines, especially subunit platforms, are discussed from immunological perspectives based on several unusual ASFV characteristics shared with HIV and poxviruses. These characteristics, including multiple distinct infectious virions, extremely high glycosylation and low antigen surface density of envelope proteins, immune evasion, and possible apoptotic mimicry, could pose enormous challenges to the development of ASF vaccines, especially subunit platforms designed to induce humoral immunity.
Collapse
|
11
|
Canter JA, Aponte T, Ramirez-Medina E, Pruitt S, Gladue DP, Borca MV, Zhu JJ. Serum Neutralizing and Enhancing Effects on African Swine Fever Virus Infectivity in Adherent Pig PBMC. Viruses 2022; 14:v14061249. [PMID: 35746720 PMCID: PMC9229155 DOI: 10.3390/v14061249] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 12/28/2022] Open
Abstract
African swine fever virus (ASFV) causes hemorrhagic fever with mortality rates of up to 100% in domestic pigs. Currently, there are no commercial vaccines for the disease. Only some live-attenuated viruses have been able to protect pigs from ASFV infection. The immune mechanisms involved in the protection are unclear. Immune sera can neutralize ASFV but incompletely. The mechanisms involved are not fully understood. Currently, there is no standardized protocol for ASFV neutralization assays. In this study, a flow cytometry-based ASFV neutralization assay was developed and tested in pig adherent PBMC using a virulent ASFV containing a fluorescent protein gene as a substrate for neutralization. As with previous studies, the percentage of infected macrophages was approximately five time higher than that of infected monocytes, and nearly all infected cells displayed no staining with anti-CD16 antibodies. Sera from naïve pigs and pigs immunized with a live-attenuated ASFV and fully protected against parental virus were used in the assay. The sera displayed incomplete neutralization with MOI-dependent neutralizing efficacies. Extracellular, but not intracellular, virions suspended in naïve serum were more infectious than those in the culture medium, as reported for some enveloped viruses, suggesting a novel mechanism of ASFV infection in macrophages. Both the intracellular and extracellular virions could not be completely neutralized.
Collapse
Affiliation(s)
- Jessica A. Canter
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Plum Island Animal Disease Center, Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Theresa Aponte
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Plum Island Animal Disease Center, Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Elizabeth Ramirez-Medina
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
| | - Sarah Pruitt
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
| | - Douglas P. Gladue
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Correspondence: (D.P.G.); (M.V.B.); (J.J.Z.); Tel.: +1-631-323-3131 (D.P.G.); +1-631-323-3035 (M.V.B.); +1-631-323-3186 (J.J.Z.)
| | - Manuel V. Borca
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Correspondence: (D.P.G.); (M.V.B.); (J.J.Z.); Tel.: +1-631-323-3131 (D.P.G.); +1-631-323-3035 (M.V.B.); +1-631-323-3186 (J.J.Z.)
| | - James J. Zhu
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Correspondence: (D.P.G.); (M.V.B.); (J.J.Z.); Tel.: +1-631-323-3131 (D.P.G.); +1-631-323-3035 (M.V.B.); +1-631-323-3186 (J.J.Z.)
| |
Collapse
|
12
|
Kennedy RB, Ovsyannikova IG, Haralambieva IH, Grill DE, Poland GA. Proteomic assessment of humoral immune responses in smallpox vaccine recipients. Vaccine 2022; 40:789-797. [PMID: 34952760 PMCID: PMC8792332 DOI: 10.1016/j.vaccine.2021.12.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 11/09/2021] [Accepted: 12/13/2021] [Indexed: 02/02/2023]
Abstract
The availability of effective smallpox vaccines was a critical element of the successful eradication of smallpox in 1980. Antibody responses play a primary role in protective immunity and neutralizing antibody is an established correlate of protection against smallpox. In this study we used a poxvirus proteome array to assess the antibody response to individual viral proteins in a cohort of 1,037 smallpox vaccine recipients. Several statistically significant differences were observed in the antibody response to immunodominant proteins between men and women, including B5R-a major target of neutralizing antibody in vaccinia immune globulin, and the membrane proteins D8L and A27L, both of which have been used as vaccine antigens providing protection in animal models. We also noted differences across racial/ethnic groups. In this cohort, which consisted of both ACAM2000 and Dryvax recipients, we noted minute differences in the antibody responses to a restricted number of viral proteins, providing additional support for the use of ACAM2000 as a replacement smallpox vaccine. Furthermore, our data indicate that poxvirus proteome microarrays can be valuable for screening and monitoring smallpox vaccine-induced humoral immune responses in large-scale serologic surveillance studies and prove useful in the guidance of developing novel smallpox candidate vaccines.
Collapse
Affiliation(s)
- Richard B. Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN USA,Department of Internal Medicine, Mayo Clinic, Rochester, MN USA,Corresponding author: Richard B. Kennedy, Ph.D., Co-Director, Mayo Vaccine Research Group, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, Phone: (507) 284-0708, Fax: (507) 266-4716,
| | - Inna G. Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN USA,Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Iana H. Haralambieva
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN USA,Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Diane E. Grill
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN USA
| | - Gregory A. Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN USA,Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
13
|
Ahsendorf HP, Diesterbeck US, Hotop SK, Winkler M, Brönstrup M, Czerny CP. Characterisation of an Anti-Vaccinia Virus F13 Single Chain Fragment Variable from a Human Anti-Vaccinia Virus-Specific Recombinant Immunoglobulin Library. Viruses 2022; 14:v14020197. [PMID: 35215792 PMCID: PMC8879190 DOI: 10.3390/v14020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 11/30/2022] Open
Abstract
Vaccinia virus (VACV) belongs to the genus Orthopoxvirus of the family Poxviridae. There are four different forms of infectious virus particles: intracellular mature virus (IMV), intracellular en-veloped virus (IEV), cell-associated enveloped virus (CEV) and extracellular enveloped virus (EEV). The F13 protein occupies the inner side of the CEV- and EEV-membranes and the outer side of the IEV-membranes. It plays an important role in wrapping progress and EEV production. We constructed a human single-chain fragment variable (scFv) library with a diversity of ≥4 × 108 independent colonies using peripheral blood from four vaccinated donors. One anti-F13 scFv was isolated and characterised after three rounds of panning. In Western blotting assays, the scFv 3E2 reacted with the recombinant F13VACV protein with a reduction of binding under denatured and reduced conditions. Two antigenic binding sites (139-GSIHTIKTLGVYSDY-153 and 169-AFNSAKNSWLNL-188) of scFv 3E2 were mapped using a cellulose membrane encompassing 372 15-mere peptides with 12 overlaps covering the whole F13 protein. No neutralisation capa-bilities were observed either in the presence or absence of complement. In conclusion, the con-struction of recombinant immunoglobulin libraries is a promising strategy to isolate specific scFvs to enable the study of the host-pathogen interaction.
Collapse
Affiliation(s)
- Henrike P. Ahsendorf
- Division of Microbiology and Animal Hygiene, Department of Animal Sciences, University of Göttingen, Burckhardtweg 2, 37077 Göttingen, Germany; (H.P.A.); (C.-P.C.)
| | - Ulrike S. Diesterbeck
- Division of Microbiology and Animal Hygiene, Department of Animal Sciences, University of Göttingen, Burckhardtweg 2, 37077 Göttingen, Germany; (H.P.A.); (C.-P.C.)
- Correspondence:
| | - Sven-Kevin Hotop
- Helmholtz Centre for Infection Research, Inhoffenstraβe 7, 38124 Braunschweig, Germany; (S.-K.H.); (M.B.)
| | - Michael Winkler
- Infection Biology Unit, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany;
| | - Mark Brönstrup
- Helmholtz Centre for Infection Research, Inhoffenstraβe 7, 38124 Braunschweig, Germany; (S.-K.H.); (M.B.)
| | - Claus-Peter Czerny
- Division of Microbiology and Animal Hygiene, Department of Animal Sciences, University of Göttingen, Burckhardtweg 2, 37077 Göttingen, Germany; (H.P.A.); (C.-P.C.)
| |
Collapse
|
14
|
Monticelli SR, Bryk P, Brewer MG, Aguilar HC, Norbury CC, Ward BM. An increase in glycoprotein concentration on extracellular virions dramatically alters vaccinia virus infectivity and pathogenesis without impacting immunogenicity. PLoS Pathog 2021; 17:e1010177. [PMID: 34962975 PMCID: PMC8746760 DOI: 10.1371/journal.ppat.1010177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 01/10/2022] [Accepted: 12/02/2021] [Indexed: 11/20/2022] Open
Abstract
The extracellular virion (EV) form of Orthopoxviruses is required for cell-to-cell spread and pathogenesis, and is the target of neutralizing antibodies in the protective immune response. EV have a double envelope that contains several unique proteins that are involved in its intracellular envelopment and/or subsequent infectivity. One of these, F13, is involved in both EV formation and infectivity. Here, we report that replacement of vaccinia virus F13L with the molluscum contagiosum virus homolog, MC021L, results in the production of EV particles with significantly increased levels of EV glycoproteins, which correlate with a small plaque phenotype. Using a novel fluorescence-activated virion sorting assay to isolate EV populations based on glycoprotein content we determine that EV containing either higher or lower levels of glycoproteins are less infectious, suggesting that there is an optimal concentration of glycoproteins in the outer envelope that is required for maximal infectivity of EV. This optimal glycoprotein concentration was required for lethality and induction of pathology in a cutaneous model of animal infection, but was not required for induction of a protective immune response. Therefore, our results demonstrate that there is a sensitive balance between glycoprotein incorporation, infectivity, and pathogenesis, and that manipulation of EV glycoprotein levels can produce vaccine vectors in which pathologic side effects are attenuated without a marked diminution in induction of protective immunity.
Collapse
Affiliation(s)
- Stephanie R. Monticelli
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Peter Bryk
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Matthew G. Brewer
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Hector C. Aguilar
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| | - Christopher C. Norbury
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Brian M. Ward
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| |
Collapse
|
15
|
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
|
16
|
Pelin A, Boulton S, Tamming LA, Bell JC, Singaravelu R. Engineering vaccinia virus as an immunotherapeutic battleship to overcome tumor heterogeneity. Expert Opin Biol Ther 2020; 20:1083-1097. [PMID: 32297534 DOI: 10.1080/14712598.2020.1757066] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Immunotherapy is a rapidly evolving area of cancer therapeutics aimed at driving a systemic immune response to fight cancer. Oncolytic viruses (OVs) are at the cutting-edge of innovation in the immunotherapy field. Successful OV platforms must be effective in reshaping the tumor microenvironment and controlling tumor burden, but also be highly specific to avoid off-target side effects. Large DNA viruses, like vaccinia virus (VACV), have a large coding capacity, enabling the encoding of multiple immunostimulatory transgenes to reshape the tumor immune microenvironment. VACV-based OVs have shown promising results in both pre-clinical and clinical studies, including safe and efficient intravenous delivery to metastatic tumors. AREA COVERED This review summarizes attenuation strategies to generate a recombinant VACV with optimal tumor selectivity and immunogenicity. In addition, we discuss immunomodulatory transgenes that have been introduced into VACV and summarize their effectiveness in controlling tumor burden. EXPERT OPINION VACV encodes several immunomodulatory genes which aid the virus in overcoming innate and adaptive immune responses. Strategic deletion of these virulence factors will enable an optimal balance between viral persistence and immunogenicity, robust tumor-specific expression of payloads and promotion of a systemic anti-cancer immune response. Rational selection of therapeutic transgenes will maximize the efficacy of OVs and their synergy in combinatorial immunotherapy schemes.
Collapse
Affiliation(s)
- Adrian Pelin
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - Stephen Boulton
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - Levi A Tamming
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - John C Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| | - Ragunath Singaravelu
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute , Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa , Ottawa, Ontario, Canada
| |
Collapse
|
17
|
Mucker EM, Lindquist M, Hooper JW. Particle-specific neutralizing activity of a monoclonal antibody targeting the poxvirus A33 protein reveals differences between cell associated and extracellular enveloped virions. Virology 2020; 544:42-54. [PMID: 32174513 DOI: 10.1016/j.virol.2020.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 10/24/2022]
Abstract
Only a small subset of the hundreds of proteins encoded by the poxvirus genome have been shown to be effective as vaccine and/or therapeutic targets. One of these proteins is A33. Here we assess and dissect the ability of an anti-A33 humanized monoclonal antibody, c6C, to affect vaccinia virus infection in vitro. Enveloped virions (EV) released from infected cells can be sensitive or resistant to neutralization by c6C indicating there are different types of EV particles, extracellular enveloped virions (EEV) and released cellular-associated virions (rCEV), that are biologically distinct. Through a combination of plaque phenotype, confocal imaging, and neutralization assays, we found that c6C differentially affects EV from two different virus strains, IHD-J and WR. Evidence for an anti-A33 resistant EV particle, and strain differences in this phenotype, provides a logical answer as to why certain functional assays in the literature have been unable to detect anti-viral effects of anti-A33 antibodies.
Collapse
Affiliation(s)
- Eric M Mucker
- Molecular Virology Branch, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, 21702, MD, USA
| | - Michael Lindquist
- Molecular Virology Branch, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, 21702, MD, USA
| | - Jay W Hooper
- Molecular Virology Branch, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, 21702, MD, USA.
| |
Collapse
|
18
|
Nakatake M, Kurosaki H, Kuwano N, Horita K, Ito M, Kono H, Okamura T, Hasegawa K, Yasutomi Y, Nakamura T. Partial Deletion of Glycoprotein B5R Enhances Vaccinia Virus Neutralization Escape while Preserving Oncolytic Function. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:159-171. [PMID: 31236440 PMCID: PMC6580015 DOI: 10.1016/j.omto.2019.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 05/09/2019] [Indexed: 11/12/2022]
Abstract
Vaccinia virus (VV) has been utilized in oncolytic virotherapy, but it risks a host antiviral immune response. VV has an extracellular enveloped virus (EEV) form consisting of a normal virion covered with a host-derived outer membrane that enables its spread via circulation while evading host immune mechanisms. However, the immune resistance of EEV is only partial, owing to expression of the surface protein B5R, which has four short consensus repeat (SCR) domains that are targeted by host immune factors. To engineer a more effective virus for oncolytic virotherapy, we developed an enhanced immune-evading oncolytic VV by removing the SCRs from the attenuated strain LC16mO. Although deletion of only the SCRs preserved viral replication, progeny production, and oncolytic activity, deletion of whole B5R led to attenuation of the virus. Importantly, SCR-deleted EEV had higher neutralization resistance than did B5R-wild-type EEV against VV-immunized animal serum; moreover, it retained oncolytic function, thereby prolonging the survival of tumor-bearing mice treated with anti-VV antibody. These results demonstrate that partial SCR deletion increases neutralization escape without affecting the oncolytic potency of VV, making it useful for the treatment of tumors under the anti-virus antibody existence.
Collapse
Affiliation(s)
- Motomu Nakatake
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Hajime Kurosaki
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Nozomi Kuwano
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Kosuke Horita
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Mai Ito
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Hiromichi Kono
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Tomotaka Okamura
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki 305-0843, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka-City, Saitama 350-1298, Japan
| | - Yasuhiro Yasutomi
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki 305-0843, Japan
| | - Takafumi Nakamura
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| |
Collapse
|
19
|
Abstract
The varied landscape of the adaptive immune response is determined by the peptides presented by immune cells, derived from viral or microbial pathogens or cancerous cells. The study of immune biomarkers or antigens is not new, and classical methods such as agglutination, enzyme-linked immunosorbent assay, or Western blotting have been used for many years to study the immune response to vaccination or disease. However, in many of these traditional techniques, protein or peptide identification has often been the bottleneck. Recent progress in genomics and mass spectrometry have led to many of the rapid advances in proteomics approaches. Immunoproteomics describes a rapidly growing collection of approaches that have the common goal of identifying and measuring antigenic peptides or proteins. This includes gel-based, array-based, mass spectrometry-based, DNA-based, or in silico approaches. Immunoproteomics is yielding an understanding of disease and disease progression, vaccine candidates, and biomarkers. This review gives an overview of immunoproteomics and closely related technologies that are used to define the full set of protein antigens targeted by the immune system during disease.
Collapse
Affiliation(s)
- Kelly M Fulton
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON, Canada
| | - Isabel Baltat
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON, Canada
| | - Susan M Twine
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON, Canada.
| |
Collapse
|
20
|
Avril A. Therapeutic Antibodies for Biodefense. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1053:173-205. [PMID: 29549640 DOI: 10.1007/978-3-319-72077-7_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Diseases can be caused naturally by biological agents such as bacteria, viruses and toxins (natural risk). However, such biological agents can be intentionally disseminated in the environment by a State (military context) or terrorists to cause diseases in a population or livestock, to destabilize a nation by creating a climate of terror, destabilizing the economy and undermining institutions. Biological agents can be classified according to the severity of illness they cause, its mortality and how easily the agent can be spread. The Centers for Diseases Control and Prevention (CDC) classify biological agents in three categories (A, B and C); Category A consists of the six pathogens most suitable for use as bioweapons (Bacillus anthracis, Yersinia pestis, Francisella tularensis, botulinum neurotoxins, smallpox and viral hemorrhagic fevers). Antibodies represent a perfect biomedical countermeasure as they present both prophylactic and therapeutic properties, act fast and are highly specific to the target. This review focuses on the main biological agents that could be used as bioweapons, the history of biowarfare and antibodies that have been developed to neutralize these agents.
Collapse
Affiliation(s)
- Arnaud Avril
- Département des maladies infectieuses, Unité biothérapies anti-infectieuses et immunité, Institut de Recherche Biomédical des Armées, Brétigny-sur-Orge, France.
| |
Collapse
|
21
|
Brown EP, Dowell KG, Boesch AW, Normandin E, Mahan AE, Chu T, Barouch DH, Bailey-Kellogg C, Alter G, Ackerman ME. Multiplexed Fc array for evaluation of antigen-specific antibody effector profiles. J Immunol Methods 2017; 443:33-44. [PMID: 28163018 PMCID: PMC5333794 DOI: 10.1016/j.jim.2017.01.010] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/27/2017] [Accepted: 01/31/2017] [Indexed: 01/09/2023]
Abstract
Antibodies are widely considered to be a frequent primary and often mechanistic correlate of protection of approved vaccines; thus evaluating the antibody response is of critical importance in attempting to understand and predict the efficacy of novel vaccine candidates. Historically, antibody responses have been analyzed by determining the titer of the humoral response using measurements such as an ELISA, neutralization, or agglutination assays. In the simplest case, sufficiently high titers of antibody against vaccine antigen(s) are sufficient to predict protection. However, antibody titer provides only a partial measure of antibody function, which is dependent on both the variable region (Fv) to bind the antigen target, and the constant region (Fc) to elicit an effector response from the innate arm of the immune system. In the case of some diseases, such as HIV, for which an effective vaccine has proven elusive, antibody effector function has been shown to be an important driver of monoclonal antibody therapy outcomes, of viral control in infected patients, and of vaccine-mediated protection in preclinical and clinical studies. We sought to establish a platform for the evaluation of the Fc domain characteristics of antigen-specific antibodies present in polyclonal samples in order to better develop insights into Fc receptor-mediated antibody effector activity, more fully understand how antibody responses may differ in association with disease progression and between subject groups, and differentiate protective from non-protective responses. To this end we have developed a high throughput biophysical platform capable of simultaneously evaluating many dimensions of the antibody effector response. High-throughput array-based characterization platform for polyclonal antibodies. Development of a biophysical proxy for antibody effector function. Antigen and Fc receptor recognition characteristics are captured. Enables systematic serologic studies of NHP and human antibody samples.
Collapse
Affiliation(s)
- Eric P Brown
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Karen G Dowell
- Department of Computer Science, Dartmouth College, Hanover, NH 03755, USA
| | - Austin W Boesch
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Erica Normandin
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Alison E Mahan
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Thach Chu
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Dan H Barouch
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | | |
Collapse
|
22
|
Modulating Antibody Functionality in Infectious Disease and Vaccination. Trends Mol Med 2016; 22:969-982. [PMID: 27756530 DOI: 10.1016/j.molmed.2016.09.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/06/2016] [Accepted: 09/11/2016] [Indexed: 12/23/2022]
Abstract
Induction of pathogen-specific binding antibodies has long been considered a signature of protective immunity following vaccination and infection. The humoral immune response is a complex network of antibodies that target different specificities and drive different functions, collectively acting to limit and clear infection either directly, via pathogen neutralization, or indirectly, via pathogen clearance by the innate immune system. Emerging data suggest that not all antibody responses are equal, and qualitative features of antibodies may be key to defining protective immune profiles. Here, we review the most recent advances in our understanding of protective functional antibody responses in natural infection, vaccination, and monoclonal antibody therapeutics. Moreover, we highlight opportunities to augment or modulate antibody-mediated protection through enhancement of antibody functionality.
Collapse
|
23
|
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
|
24
|
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
|
25
|
Brown EP, Normandin E, Osei-Owusu NY, Mahan AE, Chan YN, Lai JI, Vaccari M, Rao M, Franchini G, Alter G, Ackerman ME. Microscale purification of antigen-specific antibodies. J Immunol Methods 2015; 425:27-36. [PMID: 26078040 DOI: 10.1016/j.jim.2015.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 06/05/2015] [Accepted: 06/08/2015] [Indexed: 10/23/2022]
Abstract
Glycosylation of the Fc domain is an important driver of antibody effector function. While assessment of antibody glycoform compositions observed across total plasma IgG has identified differences associated with a variety of clinical conditions, in many cases it is the glycosylation state of only antibodies against a specific antigen or set of antigens that may be of interest, for example, in defining the potential effector function of antibodies produced during disease or after vaccination. Historically, glycoprofiling such antigen-specific antibodies in clinical samples has been challenging due to their low prevalence, the high sample requirement for most methods of glycan determination, and the lack of high-throughput purification methods. New methods of glycoprofiling with lower sample requirements and higher throughput have motivated the development of microscale and automatable methods for purification of antigen-specific antibodies from polyclonal sources such as clinical serum samples. In this work, we present a robot-compatible 96-well plate-based method for purification of antigen-specific antibodies, suitable for such population level glycosylation screening. We demonstrate the utility of this method across multiple antibody sources, using both purified plasma IgG and plasma, and across multiple different antigen types, with enrichment factors greater than 1000-fold observed. Using an on-column IdeS protease treatment, we further describe staged release of Fc and Fab domains, allowing for glycoprofiling of each domain.
Collapse
Affiliation(s)
- Eric P Brown
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Erica Normandin
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Nana Yaw Osei-Owusu
- Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH 03755, United States
| | - Alison E Mahan
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA 02139, United States
| | - Ying N Chan
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Jennifer I Lai
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Monica Vaccari
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, MD 20814, United States
| | - Mangala Rao
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States
| | - Genoveffa Franchini
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, MD 20814, United States
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA 02139, United States
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States.
| |
Collapse
|
26
|
Overton ET, Stapleton J, Frank I, Hassler S, Goepfert PA, Barker D, Wagner E, von Krempelhuber A, Virgin G, Meyer TP, Müller J, Bädeker N, Grünert R, Young P, Rösch S, Maclennan J, Arndtz-Wiedemann N, Chaplin P. Safety and Immunogenicity of Modified Vaccinia Ankara-Bavarian Nordic Smallpox Vaccine in Vaccinia-Naive and Experienced Human Immunodeficiency Virus-Infected Individuals: An Open-Label, Controlled Clinical Phase II Trial. Open Forum Infect Dis 2015; 2:ofv040. [PMID: 26380340 PMCID: PMC4567089 DOI: 10.1093/ofid/ofv040] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 03/03/2015] [Indexed: 12/13/2022] Open
Abstract
Background. First- and second-generation smallpox vaccines are contraindicated in individuals infected with human immunodeficiency virus (HIV). A new smallpox vaccine is needed to protect this population in the context of biodefense preparedness. The focus of this study was to compare the safety and immunogenicity of a replication-deficient, highly attenuated smallpox vaccine modified vaccinia Ankara (MVA) in HIV-infected and healthy subjects. Methods. An open-label, controlled Phase II trial was conducted at 36 centers in the United States and Puerto Rico for HIV-infected and healthy subjects. Subjects received 2 doses of MVA administered 4 weeks apart. Safety was evaluated by assessment of adverse events, focused physical exams, electrocardiogram recordings, and safety laboratories. Immune responses were assessed using enzyme-linked immunosorbent assay (ELISA) and a plaque reduction neutralization test (PRNT). Results. Five hundred seventy-nine subjects were vaccinated at least once and had data available for analysis. Rates of ELISA seropositivity were comparably high in vaccinia-naive healthy and HIV-infected subjects, whereas PRNT seropositivity rates were higher in healthy compared with HIV-infected subjects. Modified vaccinia Ankara was safe and well tolerated with no adverse impact on viral load or CD4 counts. There were no cases of myo-/pericarditis reported. Conclusions. Modified vaccinia Ankara was safe and immunogenic in subjects infected with HIV and represents a promising smallpox vaccine candidate for use in immunocompromised populations.
Collapse
Affiliation(s)
- Edgar Turner Overton
- Division of Infectious Diseases, University of Alabama at Birmingham, School of Medicine , Birmingham, Alabama
| | - Jack Stapleton
- Depts. of Internal Medicine, Microbiology & Immunology , University of Iowa , Iowa City
| | - Ian Frank
- Infectious Diseases Section , University of Pennsylvania , Philadelphia
| | | | - Paul A Goepfert
- Division of Infectious Diseases, University of Alabama at Birmingham, School of Medicine , Birmingham, Alabama
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Davies DH, Chun S, Hermanson G, Tucker JA, Jain A, Nakajima R, Pablo J, Felgner PL, Liang X. T cell antigen discovery using soluble vaccinia proteome reveals recognition of antigens with both virion and nonvirion association. THE JOURNAL OF IMMUNOLOGY 2014; 193:1812-27. [PMID: 25024392 DOI: 10.4049/jimmunol.1400663] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Vaccinia virus (VACV) is a useful model system for understanding the immune response to a complex pathogen. Proteome-wide Ab profiling studies reveal the humoral response to be strongly biased toward virion-associated Ags, and several membrane proteins induce Ab-mediated protection against VACV challenge in mice. Some studies have indicated that the CD4 response is also skewed toward proteins with virion association, whereas the CD8 response is more biased toward proteins with early expression. In this study, we have leveraged a VACV strain Western Reserve (VACV-WR) plasmid expression library, produced previously for proteome microarrays for Ab profiling, to make a solubilized full VACV-WR proteome for T cell Ag profiling. Splenocytes from VACV-WR-infected mice were assayed without prior expansion against the soluble proteome in assays for Th1 and Th2 signature cytokines. The response to infection was polarized toward a Th1 response, with the distribution of reactive T cell Ags comprising both early and late VACV proteins. Interestingly, the proportions of different functional subsets were similar to that present in the whole proteome. In contrast, the targets of Abs from the same mice were enriched for membrane and other virion components, as described previously. We conclude that a "nonbiasing" approach to T cell Ag discovery reveals a T cell Ag profile in VACV that is broader and less skewed to virion association than the Ab profile. The T cell Ag mapping method developed in the present study should be applicable to other organisms where expressible "ORFeome" libraries are also available, and it is readily scalable for larger pathogens.
Collapse
Affiliation(s)
- D Huw Davies
- Division of Infectious Diseases, School of Medicine, University of California, Irvine, Irvine, CA 92697; Antigen Discovery, Inc., Irvine, CA 92618; and
| | - Sookhee Chun
- Division of Infectious Diseases, School of Medicine, University of California, Irvine, Irvine, CA 92697
| | | | - Jo Anne Tucker
- Division of Hematology and Oncology, School of Medicine, University of California, Irvine, Irvine, CA 92697
| | - Aarti Jain
- Division of Infectious Diseases, School of Medicine, University of California, Irvine, Irvine, CA 92697
| | - Rie Nakajima
- Division of Infectious Diseases, School of Medicine, University of California, Irvine, Irvine, CA 92697
| | - Jozelyn Pablo
- Division of Infectious Diseases, School of Medicine, University of California, Irvine, Irvine, CA 92697; Antigen Discovery, Inc., Irvine, CA 92618; and
| | - Philip L Felgner
- Division of Infectious Diseases, School of Medicine, University of California, Irvine, Irvine, CA 92697
| | | |
Collapse
|
28
|
Human antibody responses to the polyclonal Dryvax vaccine for smallpox prevention can be distinguished from responses to the monoclonal replacement vaccine ACAM2000. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:877-85. [PMID: 24759651 DOI: 10.1128/cvi.00035-14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Dryvax (Wyeth Laboratories, Inc., Marietta, PA) is representative of the vaccinia virus preparations that were previously used for preventing smallpox. While Dryvax was highly effective, the national supply stocks were depleted, and there were manufacturing concerns regarding sterility and the clonal heterogeneity of the vaccine. ACAM2000 (Acambis, Inc./Sanofi-Pasteur Biologics Co., Cambridge, MA), a single-plaque-purified vaccinia virus derivative of Dryvax, recently replaced the polyclonal smallpox vaccine for use in the United States. A substantial amount of sequence heterogeneity exists within the polyclonal proteome of Dryvax, including proteins that are missing from ACAM2000. Reasoning that a detailed comparison of antibody responses to the polyclonal and monoclonal vaccines may be useful for identifying unique properties of each antibody response, we utilized a protein microarray comprised of approximately 94% of the vaccinia poxvirus proteome (245 proteins) to measure protein-specific antibody responses of 71 individuals receiving a single vaccination with ACAM2000 or Dryvax. We observed robust antibody responses to 21 poxvirus proteins in vaccinated individuals, including 11 proteins that distinguished Dryvax responses from ACAM2000. Analysis of protein sequences from Dryvax clones revealed amino acid level differences in these 11 antigenic proteins and suggested that sequence variation and clonal heterogeneity may contribute to the observed differences between Dryvax and ACAM2000 antibody responses.
Collapse
|
29
|
Abstract
The varied landscape of the adaptive immune response is determined by the peptides presented by immune cells, derived from viral or microbial pathogens or cancerous cells. The study of immune biomarkers or antigens is not new and classical methods such as agglutination, enzyme-linked immunosorbent assay, or Western blotting have been used for many years to study the immune response to vaccination or disease. However, in many of these traditional techniques, protein or peptide identification has often been the bottleneck. Recent advances in genomics and proteomics, has led to many of the rapid advances in proteomics approaches. Immunoproteomics describes a rapidly growing collection of approaches that have the common goal of identifying and measuring antigenic peptides or proteins. This includes gel based, array based, mass spectrometry, DNA based, or in silico approaches. Immunoproteomics is yielding an understanding of disease and disease progression, vaccine candidates, and biomarkers. This review gives an overview of immunoproteomics and closely related technologies that are used to define the full set of antigens targeted by the immune system during disease.
Collapse
Affiliation(s)
- Kelly M Fulton
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | | |
Collapse
|