1
|
Kobiyama K, Utsumi D, Kaku Y, Sasaki E, Yasui F, Okamura T, Onodera T, Tobuse AJ, Sakkour A, Amiry AF, Hayashi T, Temizoz B, Liu K, Negishi H, Toyama-Sorimachi N, Kohara M, Sawasaki T, Takagi J, Sato K, Takahashi Y, Yasutomi Y, Ishii KJ. Immunological analysis of LC16m8 vaccine: preclinical and early clinical insights into mpox. EBioMedicine 2025; 115:105703. [PMID: 40239465 PMCID: PMC12020844 DOI: 10.1016/j.ebiom.2025.105703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 03/19/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND The global mpox outbreak (2022-2024) highlights the need for effective and safe vaccines, particularly for vulnerable populations. The LC16m8 vaccine, an attenuated vaccinia virus strain for smallpox, shows promise in inducing immunity against the monkeypox virus (MPXV). METHODS We conducted a comprehensive immunological evaluation of LC16m8 in mice, non-human primates, and humans. FINDINGS LC16m8 induced strong humoural responses in BALB/c, C57BL/6J, and CAST/EiJ mice, targeting MPXV H3, A35, and M1R antigens, promoting germinal centre B cells and follicular helper T cells, essential for long-term immunity. Vaccinated CAST/EiJ mice showed reduced lung MPXV viral loads, demonstrating efficacy. In humans, LC16m8 enhanced neutralising antibodies against multiple MPXV clades, suggesting broad protection. In cynomolgus monkeys, systemic administration caused localised pox lesions without significantly affecting weight, temperature, or haematological parameters. INTERPRETATION This cross-species immunological analysis provides preclinical and early clinical insights into LC16m8's efficacy and safety against mpox. While LC16m8 enhanced antibody responses against MPXV clade Ia and Ib, further studies are required to evaluate its efficacy, particularly in naive and immunocompromised populations. FUNDING This research was supported by AMED under Grant Numbers JP243fa727002, JP243fa727001s0703, and JP243fa627001h0003 (K.J.I), JP24jf0126002, JP24fk0108690, JP243fa627001h0003, and JP243fa727002 (K.S), JP243fa727002 (Y.T.), JP243fa727002 and JP243fa627007h0003 (Y.Y.), and by the Research Support Project for Life Science and Drug Discovery (BINDS) from AMED under Grant Number JP23ama121011 (J.T.), and JP23ama121010 (T.S.), and by the Ministry of Education, Culture, Sports, Science and Technology in Japan under Grant Number 23K06577 (E.S.). AMED under Grant Number JP233fa827017 and JP243fa827017 (F.Y.), JP22fk0108501 (M.K.).
Collapse
Affiliation(s)
- Kouji Kobiyama
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Daichi Utsumi
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan
| | - Yu Kaku
- Division of Systems Virology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Eita Sasaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Fumihiko Yasui
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tomotaka Okamura
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Asuka Joy Tobuse
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Areej Sakkour
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ahmad Faisal Amiry
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tomoya Hayashi
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Burcu Temizoz
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kaiwen Liu
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hideo Negishi
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Noriko Toyama-Sorimachi
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Human Immunology, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tatsuya Sawasaki
- Proteo-Science Center (PROS), Ehime University, Matsuyama, Japan
| | - Junichi Takagi
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Kei Sato
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Systems Virology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan; Collaboration Unit for Infection, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan; MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yasuhiro Yasutomi
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan
| | - Ken J Ishii
- Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan; The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
2
|
Li Y, Wang L, Chen S. An overview of the progress made in research into the Mpox virus. Med Res Rev 2025; 45:788-812. [PMID: 39318037 DOI: 10.1002/med.22085] [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: 03/26/2024] [Revised: 08/05/2024] [Accepted: 09/01/2024] [Indexed: 09/26/2024]
Abstract
Mpox is a zoonotic illness caused by the Mpox virus (MPXV), a member of the Orthopoxvirus family. Although a few cases have been reported outside Africa, it was originally regarded as an endemic disease limited to African countries. However, the Mpox outbreak of 2022 was remarkable in that the infection spread to more than 123 countries worldwide, causing thousands of infections and deaths. The ongoing Mpox outbreak has been declared as a public health emergency of international concern by the World Health Organization. For a better management and control of the epidemic, this review summarizes the research advances and important scientific findings on MPXV by reviewing the current literature on epidemiology, clinical characteristics, diagnostic methods, prevention and treatment measures, and animal models of MPXV. This review provides useful information to raise awareness about the transmission, symptoms, and protective measures of MPXV, serving as a theoretical guide for relevant institutions to control MPXV.
Collapse
Affiliation(s)
- Yansheng Li
- Shenzhen Key Laboratory of Microbiology in Genomic Modification & Editing and Application, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound lmaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Department of Critical Care Medicine, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Lianrong Wang
- Department of Respiratory Diseases, Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Shi Chen
- Shenzhen Key Laboratory of Microbiology in Genomic Modification & Editing and Application, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound lmaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Department of Critical Care Medicine, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
3
|
Eslami A, Alimoghadam S, Khoshravesh S, Shirani M, Alimoghadam R, Alavi Darazam I. Mpox vaccination and treatment: a systematic review. J Chemother 2024; 36:85-109. [PMID: 38069596 DOI: 10.1080/1120009x.2023.2289270] [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: 03/17/2023] [Accepted: 11/27/2023] [Indexed: 02/01/2024]
Abstract
The Human monkeypox virus (mpox) belongs to the Poxviridae family, characterized by double-stranded DNA. A 2022 outbreak, notably prevalent among men who have sex with men, was confirmed by the World Health Organization. To understand shifting prevalence patterns and clinical manifestations, we conducted a systematic review of recent animal and human studies. We comprehensively searched PubMed, Scopus, Web of Science, Cochrane Library, and Clinicaltrials.gov, reviewing 69 relevant articles from 4,342 screened records. Our analysis highlights Modified Vaccinia Ankara - Bavarian Nordic (MVA-BN)'s potential, though efficacy concerns exist. Tecovirimat emerged as a prominent antiviral in the recent outbreak. However, limited evidence underscores the imperative for further clinical trials in understanding and managing monkeypox.
Collapse
Affiliation(s)
- Arvin Eslami
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Mahsa Shirani
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Ilad Alavi Darazam
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Infectious Diseases, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Nasrin T, Samim Hassan M, Iqbal M, Yousif A, Hoque M, Ali N, Ali S. Elucidating the evolution of monkeypox virus genomes through phylo-geo-network and haplogroup analysis. J Genet Eng Biotechnol 2024; 22:100346. [PMID: 38494259 PMCID: PMC10860881 DOI: 10.1016/j.jgeb.2023.100346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 03/19/2024]
Abstract
BACKGROUND As the world settles down from the COVID-19 pandemic, many countries are faced with an unexpected outbreak of monkeypox infection. Monkeypox is a zoonotic disease caused by monkeypox virus (MPXV), which is an enveloped, double stranded DNA virus belonging to the Poxviridae family. Presently, we construct and analyze the phylo-geo-network and the corresponding haplogroups. Presently, we performed the haplogroup analysis with their defining mutations and phylogenetic lineage study along with geographical distributions with the aim to understand the evolutionary path of the MPXV across the world. RESULTS Information about 719 full length genomes of MPXV were collected from GISAID repository and the sequences extracted from NCBI. The alignment of 719 MPXV genomes and their subsequent analysis revealed a total of 1530 segregating sites of which 330 were parsimony informative (PI) sites. The variations had a positive value of Tajima's D statistic indicating some mutations being prevalent and hence balancing selection. A total of 39 haplogroups were observed in the phylo-geo-network and their defining mutations along with the evolutionary path has been discussed. The phylo-geo-network revealed the nodal haplogroup is represented by GISAID ID 13889450, haplogroup A1, an isolate from Germany, having a total of 296 identical sequences in the study incident across 22 countries. The localized evolution is highlighted by country specific sequences and haplogroups. USA had a total of 58 genomes and 13 haplogroups as compared to Peru (89 genomes, 7 haplogroups) and Germany (26 genomes, 6 haplogroups). CONCLUSIONS The evolution of MPXV can be happening in a localized manner and hence accumulation of variations in the MPXV genomes needs to be monitored in order to be prepared for any possible threats.
Collapse
Affiliation(s)
- Taslima Nasrin
- Clinical and Applied Genomics (CAG) Laboratory, Department of Biological Sciences, Aliah University, Kolkata, India.
| | - Md Samim Hassan
- Clinical and Applied Genomics (CAG) Laboratory, Department of Biological Sciences, Aliah University, Kolkata, India
| | - Muzaffar Iqbal
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Amar Yousif
- School of Pharmacy, University of Maryland Eastern Shore, Princess Anne 21853, MD, USA.
| | - Mehboob Hoque
- Applied Biochemistry Laboratory, Department of Biological Sciences, Aliah University, Kolkata, India.
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| | - Safdar Ali
- Clinical and Applied Genomics (CAG) Laboratory, Department of Biological Sciences, Aliah University, Kolkata, India.
| |
Collapse
|
5
|
Grabenstein JD, Hacker A. Vaccines against mpox: MVA-BN and LC16m8. Expert Rev Vaccines 2024; 23:796-811. [PMID: 39188013 DOI: 10.1080/14760584.2024.2397006] [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: 05/09/2024] [Revised: 08/08/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024]
Abstract
INTRODUCTION Global outbreaks involving mpox clade IIb began in mid-2022. Today, clade IIb and clade I outbreaks continue. Reliable mpox vaccines can prevent serious mpox disease and death. AREAS COVERED Globally, two vaccines hold mpox indications, regardless of mpox viral clade: MVA-BN (Bavarian Nordic) and LC16m8 (KM Biologics). This review summarizes the human and pivotal animal data establishing safety and efficacy for MVA-BN and LC16m8, including real-world evidence gathered during mpox outbreaks from 2022 through 2024. EXPERT OPINION Some regulatory decisions for MVA-BN and LC16m8 followed pathways based on surrogate outcomes, including lethal-challenge studies in nonhuman primates, among other atypical aspects. Nonetheless, MVA-BN and LC16m8 hold unencumbered registration in multiple countries. Effectiveness of MVA-BN as primary preventive vaccination (PPV) in humans against clade IIb mpox is clear from real-world studies; effectiveness of LC16m8 against clade IIb is likely from surrogate endpoints. Effectiveness of MVA-BN and LC16m8 as PPV against more-lethal clade I is likely, based on animal-challenge studies with multiple orthopoxvirus species and other studies. Both vaccines have solid safety records. MVA-BN's replication incompetence favors adoption, whereas LC16m8 has more pediatric data. Additional real-world evidence, in additional geographic settings and special populations (e.g. pregnancy, immune suppression, atopic dermatitis), is needed.
Collapse
Affiliation(s)
| | - Adam Hacker
- Coalition for Epidemic Preparedness & Innovation, Oslo, Norway
| |
Collapse
|
6
|
Neto TAP, Sidney J, Grifoni A, Sette A. Correlative CD4 and CD8 T-cell immunodominance in humans and mice: Implications for preclinical testing. Cell Mol Immunol 2023; 20:1328-1338. [PMID: 37726420 PMCID: PMC10616275 DOI: 10.1038/s41423-023-01083-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023] Open
Abstract
Antigen-specific T-cell recognition is restricted by Major Histocompatibility Complex (MHC) molecules, and differences between CD4 and CD8 immunogenicity in humans and animal species used in preclinical vaccine testing are yet to be fully understood. In this study, we addressed this matter by analyzing experimentally identified epitopes based on published data curated in the Immune Epitopes DataBase (IEDB) database. We first analyzed SARS-CoV-2 spike (S) and nucleoprotein (N), which are two common targets of the immune response and well studied in both human and mouse systems. We observed a weak but statistically significant correlation between human and H-2b mouse T-cell responses (CD8 S specific (r = 0.206, p = 1.37 × 10-13); CD4 S specific (r = 0.118, p = 2.63 × 10-5) and N specific (r = 0.179, p = 2.55 × 10-4)). Due to intrinsic differences in MHC molecules across species, we also investigated the association between the immunodominance of common Human Leukocyte Antigen (HLA) alleles for which HLA transgenic mice are available, namely, A*02:01, B*07:02, DRB1*01:01, and DRB1*04:01, and found higher significant correlations for both CD8 and CD4 (maximum r = 0.702, p = 1.36 × 10-31 and r = 0.594, p = 3.04-122, respectively). Our results further indicated that some regions are commonly immunogenic between humans and mice (either H-2b or HLA transgenic) but that others are human specific. Finally, we noted a significant correlation between CD8 and CD4 S- (r = 0.258, p = 7.33 × 1021) and N-specific (r = 0.369, p = 2.43 × 1014) responses, suggesting that discrete protein subregions can be simultaneously recognized by T cells. These findings were confirmed in other viral systems, providing general guidance for the use of murine models to test T-cell immunogenicity of viral antigens destined for human use.
Collapse
Affiliation(s)
- Tertuliano Alves Pereira Neto
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA.
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, 92037, USA
| |
Collapse
|
7
|
Turtle L, Subramaniam K. Modified vaccinia Ankara-Bavarian Nordic vaccine against mpox in children. THE LANCET. INFECTIOUS DISEASES 2023; 23:989-990. [PMID: 37336223 PMCID: PMC10275626 DOI: 10.1016/s1473-3099(23)00345-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 06/21/2023]
Affiliation(s)
- Lance Turtle
- Institute of Infection, Veterinary and Ecological Sciences University of Liverpool, Liverpool, L69 7BE, UK; Tropical & Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust (member of Liverpool Health Partners), Liverpool, UK.
| | - Krishanthi Subramaniam
- Institute of Infection, Veterinary and Ecological Sciences University of Liverpool, Liverpool, L69 7BE, UK
| |
Collapse
|
8
|
Rabaan AA, Al-Shwaikh SA, Alfouzan WA, Al-Bahar AM, Garout M, Halwani MA, Albayat H, Almutairi NB, Alsaeed M, Alestad JH, Al-Mozaini MA, Ashgar TMA, Alotaibi S, Abuzaid AA, Aldawood Y, Alsaleh AA, Al-Afghani HM, Altowaileb JA, Alshukairi AN, Arteaga-Livias K, Singh KKB, Imran M. A Comprehensive Review on Monkeypox Viral Disease with Potential Diagnostics and Therapeutic Options. Biomedicines 2023; 11:1826. [PMID: 37509466 PMCID: PMC10376530 DOI: 10.3390/biomedicines11071826] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
The purpose of this review is to give an up-to-date, thorough, and timely overview of monkeypox (Mpox), a severe infectious viral disease. Furthermore, this review provides an up-to-date treatment option for Mpox. The monkeypox virus (MPXV) has remained the most virulent poxvirus for humans since the elimination of smallpox approximately 41 years ago, with distribution mainly in central and west Africa. Mpox in humans is a zoonotically transferred disease that results in symptoms like those of smallpox. It had spread throughout west and central Africa when it was first diagnosed in the Republic of Congo in 1970. Mpox has become a major threat to global health security, necessitating a quick response by virologists, veterinarians, public health professionals, doctors, and researchers to create high-efficiency diagnostic tests, vaccinations, antivirals, and other infection control techniques. The emergence of epidemics outside of Africa emphasizes the disease's global significance. A better understanding of Mpox's dynamic epidemiology may be attained by increased surveillance and identification of cases.
Collapse
Affiliation(s)
- Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Seham A Al-Shwaikh
- Department of Commitment Management, Directorate of Health Affairs in the Eastern Province, Dammam 31176, Saudi Arabia
| | - Wadha A Alfouzan
- Department of Microbiology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait
- Microbiology Unit, Department of Laboratories, Farwania Hospital, Farwania 85000, Kuwait
| | - Ali M Al-Bahar
- Department of Laboratory, Dhahran Long Term Care Hospital, Dhahran 34257, Saudi Arabia
| | - Mohammed Garout
- Department of Community Medicine and Health Care for Pilgrims, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Muhammad A Halwani
- Department of Medical Microbiology, Faculty of Medicine, Al Baha University, Al Baha 4781, Saudi Arabia
| | - Hawra Albayat
- Infectious Disease Department, King Saud Medical City, Riyadh 7790, Saudi Arabia
| | - Norah B Almutairi
- Infectious Disease Department, King Saud Medical City, Riyadh 7790, Saudi Arabia
| | - Mohammed Alsaeed
- Infectious Disease Division, Department of Medicine, Prince Sultan Military Medical City, Riyadh 11159, Saudi Arabia
| | - Jeehan H Alestad
- Immunology and Infectious Microbiology Department, University of Glasgow, Glasgow G1 1XQ, UK
- Microbiology Department, Collage of Medicine, Jabriya 46300, Kuwait
| | - Maha A Al-Mozaini
- Immunocompromsised Host Research Section, Department of Infection and Immunity, King Faisal, Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
| | - Tala M Al Ashgar
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Sultan Alotaibi
- Molecular Microbiology Department, King Fahad Medical City, Riyadh 11525, Saudi Arabia
| | - Abdulmonem A Abuzaid
- Medical Microbiology Department, Security Forces Hospital Programme, Dammam 32314, Saudi Arabia
| | - Yahya Aldawood
- Clinical Laboratory Science Department, Mohammed Al-Mana College for Medical Sciences, Dammam 34222, Saudi Arabia
| | - Abdulmonem A Alsaleh
- Clinical Laboratory Science Department, Mohammed Al-Mana College for Medical Sciences, Dammam 34222, Saudi Arabia
| | - Hani M Al-Afghani
- Laboratory Department, Security Forces Hospital, Makkah 24269, Saudi Arabia
- iGene Center for Research and Training, Jeddah 2022, Saudi Arabia
| | - Jaffar A Altowaileb
- Microbiology Laboratory, Laboratory Department, Qatif Central Hospital, Qatif 32654, Saudi Arabia
| | - Abeer N Alshukairi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Medicine, King Faisal Specialist Hospital and Research Center, Jeddah 22233, Saudi Arabia
| | - Kovy Arteaga-Livias
- Escuela de Medicina-Filial Ica, Universidad Privada San Juan Bautista, Ica 11000, Peru
- Escuela de Medicina, Universidad Nacional Hermilio Valdizán, Huanuco 10000, Peru
| | - Kirnpal Kaur Banga Singh
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| |
Collapse
|
9
|
Abstract
BACKGROUND In addition to the COVID-19 waves, the globe is recently facing global monkeypox (MPX) outbreak. As the daily confirmed cases of MPX infection across epidemic and nonepidemic countries are increasing, taking measures to control global pandemic remains crucial. Therefore, this review aimed to provide fundamental knowledge for the prevention and control of future outbreaks of this emerging epidemic. METHODS The review was conducted using PubMed and Google Scholar databases; the search terms used were "monkeypox," "MPX tropism," "replication signaling of MPX," "biology and pathogenicity of MPX," "diagnosis of MPX," "treatment of MPX," "prevention of MPX," etc. The update epidemic data were collected from the websites of the World Health Organization (WHO), United States Centers for Disease Control and Prevention (CDC), and Africa Center for Disease Control and Prevention (ADCC). High-quality research results published in authoritative journals were summarized and preferred cited. Excluding all duplicates, non-English published references, and irrelevant literature, totally 1,436 articles were assessed for eligibility. RESULTS It is still difficult to diagnose the patient as MPX simply based on clinical manifestations; therefore, under this situation, employing polymerase chain reaction (PCR) technology to provide confirmed evidence for the diagnosis of MPX seems to be the preferred and indispensable strategy. The treatment approach for MPX infection is mainly symptomatic and supportive; anti-smallpox virus drugs including tecovirimat, cidofovir, and brincidofovir can be employed in severe cases. Timely identification and isolation of confirmed cases, cutting off dissemination routes, and vaccination of close contacts are effective measures to control MPX. Also, smallpox vaccines (JYNNEOS, LC16m8, and ACAM2000) can be under consideration due to their immunological cross-protection among Orthopoxvirus. Nevertheless, given the low quality and scarcity of relevant evidence of current antiviral drugs and vaccines, deeply seeking for the MAPK/ERK, PAK-1, PI3K/Akt signaling, and other pathways involved in MPX invasion may provide potential targets for the treatment, prevention, and control of the epidemic. CONCLUSIONS In response to the current MPX epidemic, the development of vaccines and antiviral drugs against MPX, as well as the rapid and precise diagnostic methods are still urgently needed. Sound monitoring and detection systems should be established to limit the rapid spread of MPX worldwide.
Collapse
Affiliation(s)
- Yue Kang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yue Yu
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian China
| | - Silu Xu
- Department of Pharmacy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
10
|
Niu L, Liang D, Ling Q, Zhang J, Li Z, Zhang D, Xia P, Zhu Z, Lin J, Shi A, Ma J, Yu P, Liu X. Insights into monkeypox pathophysiology, global prevalence, clinical manifestation and treatments. Front Immunol 2023; 14:1132250. [PMID: 37026012 PMCID: PMC10070694 DOI: 10.3389/fimmu.2023.1132250] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/02/2023] [Indexed: 04/08/2023] Open
Abstract
On 23rd July 2022, the World Health Organization (WHO) recognized the ongoing monkeypox outbreak as a public medical crisis. Monkeypox virus (MPV), the etiological agent of monkeypox, is a zoonotic, linear, double-stranded DNA virus. In 1970, the Democratic Republic of the Congo reported the first case of MPV infection. Human-to-human transmission can happen through sexual contact, inhaled droplets, or skin-to-skin contact. Once inoculated, the viruses multiply rapidly and spread into the bloodstream to cause viremia, which then affect multiple organs, including the skin, gastrointestinal tract, genitals, lungs, and liver. By September 9, 2022, more than 57,000 cases had been reported in 103 locations, especially in Europe and the United States. Infected patients are characterized by physical symptoms such as red rash, fatigue, backache, muscle aches, headache, and fever. A variety of medical strategies are available for orthopoxviruses, including monkeypox. Monkeypox prevention following the smallpox vaccine has shown up to 85% efficacy, and several antiviral drugs, such as Cidofovir and Brincidofovir, may slow the viral spread. In this article, we review the origin, pathophysiology, global epidemiology, clinical manifestation, and possible treatments of MPV to prevent the propagation of the virus and provide cues to generate specific drugs.
Collapse
Affiliation(s)
- Liyan Niu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Dingfa Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Queen Mary College of Nanchang University, Nanchang, China
| | - Qin Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jing Zhang
- Queen Mary College of Nanchang University, Nanchang, China
| | - Ziwen Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Deju Zhang
- Third Department of Internal Medicine, Dexing Hospital of Traditional Chinese Medicine, Dexing, Jiangxi, China
| | - Panpan Xia
- Third Department of Internal Medicine, Dexing Hospital of Traditional Chinese Medicine, Dexing, Jiangxi, China
| | - Zicheng Zhu
- Third Department of Internal Medicine, Dexing Hospital of Traditional Chinese Medicine, Dexing, Jiangxi, China
| | - Jitao Lin
- Third Department of Internal Medicine, Dexing Hospital of Traditional Chinese Medicine, Dexing, Jiangxi, China
| | - Ao Shi
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- School of Medicine, St. George University of London, London, United Kingdom
| | - Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Peng Yu
- Third Department of Internal Medicine, Dexing Hospital of Traditional Chinese Medicine, Dexing, Jiangxi, China
| | - Xiao Liu
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
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
|
12
|
Poland GA, Kennedy RB, Tosh PK. Prevention of monkeypox with vaccines: a rapid review. THE LANCET. INFECTIOUS DISEASES 2022; 22:e349-e358. [PMID: 36116460 PMCID: PMC9628950 DOI: 10.1016/s1473-3099(22)00574-6] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 01/09/2023]
Abstract
The largest outbreak of monkeypox in history began in May, 2022, and has rapidly spread across the globe ever since. The purpose of this Review is to briefly describe human immune responses to orthopoxviruses; provide an overview of the vaccines available to combat this outbreak; and discuss the various clinical data and animal studies evaluating protective immunity to monkeypox elicited by vaccinia virus-based smallpox vaccines, address ongoing concerns regarding the outbreak, and provide suggestions for the appropriate use of vaccines as an outbreak control measure. Data showing clinical effectiveness (~85%) of smallpox vaccines against monkeypox come from surveillance studies conducted in central Africa in the 1980s and later during outbreaks in the same area. These data are supported by a large number of animal studies (primarily in non-human primates) with live virus challenge by various inoculation routes. These studies uniformly showed a high degree of protection and immunity against monkeypox virus following vaccination with various smallpox vaccines. Smallpox vaccines represent an effective countermeasure that can be used to control monkeypox outbreaks. However, smallpox vaccines do cause side-effects and the replication-competent, second-generation vaccines have contraindications. Third-generation vaccines, although safer for use in immunocompromised populations, require two doses, which is an impediment to rapid outbreak response. Lessons learned from the COVID-19 pandemic should be used to inform our collective response to this monkeypox outbreak and to future outbreaks.
Collapse
Affiliation(s)
| | | | - Pritish K Tosh
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, MN, USA,Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
13
|
Domán M, Fehér E, Varga-Kugler R, Jakab F, Bányai K. Animal Models Used in Monkeypox Research. Microorganisms 2022; 10:2192. [PMID: 36363786 PMCID: PMC9694439 DOI: 10.3390/microorganisms10112192] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/21/2022] [Accepted: 10/28/2022] [Indexed: 07/26/2023] Open
Abstract
Monkeypox is an emerging zoonotic disease with a growing prevalence outside of its endemic area, posing a significant threat to public health. Despite the epidemiological and field investigations of monkeypox, little is known about its maintenance in natural reservoirs, biological implications or disease management. African rodents are considered possible reservoirs, although many mammalian species have been naturally infected with the monkeypox virus (MPXV). The involvement of domestic livestock and pets in spillover events cannot be ruled out, which may facilitate secondary virus transmission to humans. Investigation of MPXV infection in putative reservoir species and non-human primates experimentally uncovered novel findings relevant to the course of pathogenesis, virulence factors and transmission of MPXV that provided valuable information for designing appropriate prevention measures and effective vaccines.
Collapse
Affiliation(s)
- Marianna Domán
- Veterinary Medical Research Institute, H-1143 Budapest, Hungary
| | - Enikő Fehér
- Veterinary Medical Research Institute, H-1143 Budapest, Hungary
| | | | - Ferenc Jakab
- National Laboratory of Virology, Virological Research Group, Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Krisztián Bányai
- Veterinary Medical Research Institute, H-1143 Budapest, Hungary
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, H-1078 Budapest, Hungary
| |
Collapse
|
14
|
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: 95] [Impact Index Per Article: 31.7] [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
|
15
|
Quach HQ, Ovsyannikova IG, Poland GA, Kennedy RB. Evaluating immunogenicity of pathogen-derived T-cell epitopes to design a peptide-based smallpox vaccine. Sci Rep 2022; 12:15401. [PMID: 36100624 PMCID: PMC9470075 DOI: 10.1038/s41598-022-19679-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022] Open
Abstract
Despite the eradication in 1980, developing safe and effective smallpox vaccines remains an active area of research due to the recent outbreaks and the public health concern that smallpox viruses could be used as bioterrorism weapons. Identifying immunogenic peptides (epitopes) would create a foundation for the development of a robust peptide-based vaccine. We previously identified a library of naturally-processed, human leukocyte antigen class I-presented vaccinia-derived peptides from infected B cells. In the current study, we evaluated the immunogenicity of these T-cell peptides in both transgenic mouse models and human peripheral blood mononuclear cells. A vaccine based on four selected peptides provided 100% protection against a lethal viral challenge. In addition, responses from memory T cells remained unchanged up to five months. Our results validate a practical approach for identifying and verifying immunogenic peptides for vaccine development and highlight the potential of peptide-based vaccines for various infectious diseases.
Collapse
Affiliation(s)
- Huy Quang Quach
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
16
|
Mucker EM, Shamblin JD, Raymond JL, Twenhafel NA, Garry RF, Hensley LE. Effect of Monkeypox Virus Preparation on the Lethality of the Intravenous Cynomolgus Macaque Model. Viruses 2022; 14:1741. [PMID: 36016363 PMCID: PMC9413320 DOI: 10.3390/v14081741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 02/05/2023] Open
Abstract
For over two decades, researchers have sought to improve smallpox vaccines and also develop therapies to ensure protection against smallpox or smallpox-like disease. The 2022 human monkeypox pandemic is a reminder that these efforts should persist. Advancing such therapies have involved animal models primarily using surrogate viruses such as monkeypox virus. The intravenous monkeypox model in macaques produces a disease that is clinically similar to the lesional phase of fulminant human monkeypox or smallpox. Two criticisms of the model have been the unnatural route of virus administration and the high dose required to induce severe disease. Here, we purified monkeypox virus with the goal of lowering the challenge dose by removing cellular and viral contaminants within the inoculum. We found that there are advantages to using unpurified material for intravenous exposures.
Collapse
Affiliation(s)
- Eric M. Mucker
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Josh D. Shamblin
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Jo Lynne Raymond
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Nancy A. Twenhafel
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Robert F. Garry
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
- Zalgen Labs, Frederick, MD 21703, USA
- Global Virus Network (GVN), Baltimore, MD 21201, USA
| | - Lisa E. Hensley
- Zoonotic and Emerging Disease Unit, United States Department of Agriculture, Manhattan, KS 66505, USA
| |
Collapse
|
17
|
Hraib M, Jouni S, Albitar MM, Alaidi S, Alshehabi Z. The outbreak of monkeypox 2022: An overview. Ann Med Surg (Lond) 2022; 79:104069. [PMID: 35860140 PMCID: PMC9289401 DOI: 10.1016/j.amsu.2022.104069] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/22/2022] [Indexed: 12/17/2022] Open
Abstract
On May 6, 2022 an outbreak of monkeypox (MPX) was confirmed in the United Kingdom, originating from a British resident who had travelled to Nigeria. As of May 21, 2022, 92 cases have been confirmed worldwide, from 13 countries where monkeypox virus (MPXV) is not endemic. Reported cases thus far have mainly but not exclusively been identified among gay and bisexual men aged 20–50. MPXV is a viral zoonosis transmitted to humans via contacting or eating an infected animal, and direct connect with natural host's blood and body fluids. In addition to contacting with a patient's respiratory droplets, lesions, body fluids and polluted personal objects. Symptoms including shivers, headaches, fainting, backaches, and myodynia do not have any specific characteristics making it difficult to establish a proper diagnosis. Nevertheless, lymphatic hyperplasia, one of the most common symptoms of monkeypox, can be useful for diagnosing the disease. Clinical symptoms help establish the suspicion of monkeypox. However, in the absence of confirmed diagnostic tests it is very difficult to verify the disease and determine its cause based on clinical symptoms alone. There are numerous methods for detecting MPX, involving genetic, phenotypic, immunological methods, and electron microscopy. These tests require modern equipment and expert hands, which may not be available in developing countries where this disease is prevalent. Currently, there is no definite treatment for MPX. CDC recommends administering the smallpox vaccine within 4 days of exposure which may prevent the disease from happening, and within 2 weeks to reduce symptoms severity. To promptly identify patients and prevent further spreading, physicians should be aware of the travel or contact history of the patient with compatible symptoms. On May 6, 2022 an outbreak of monkeypox (MPX) was confirmed in the United Kingdom, originating from a British resident who had travelled to Nigeria. As of May 21, 2022, 92 cases have been confirmed worldwide, from 13 countries where monkeypox virus (MPXV) is not endemic. MPXV is a viral zoonosis transmitted to humans via contacting or eating an infected animal, and direct connect with natural host's blood and body fluids. In addition to contacting with a patient's respiratory droplets, lesions, body fluids and polluted personal objects. Symptoms do not have any specific characteristics. Nevertheless, lymphatic hyperplasia, one of the most common symptoms of monkeypox, can be useful for diagnosing the disease.
Collapse
Affiliation(s)
- Munawar Hraib
- Faculty of Medicine, Tishreen University, Latakia, Syria
| | - Sarah Jouni
- Faculty of Medicine, Tishreen University, Latakia, Syria
| | | | - Sara Alaidi
- Faculty of Medicine, Tishreen University, Latakia, Syria
| | - Zuheir Alshehabi
- Department of Pathology, Tishreen University Hospital, Latakia, Syria
| |
Collapse
|
18
|
Lim H, In HJ, Kim YJ, Jang S, Lee YH, Kim SH, Lee SH, Park JH, Yang HJ, Yoo JS, Lee SW, Kim MY, Chung GT, Yeo SG. Development of an attenuated smallpox vaccine candidate: The KVAC103 strain. Vaccine 2021; 39:5214-5223. [PMID: 34334254 DOI: 10.1016/j.vaccine.2021.06.060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 05/14/2021] [Accepted: 06/22/2021] [Indexed: 11/29/2022]
Abstract
Smallpox, a disease caused by the variola virus, is one of the most dangerous diseases and had killed numerous people before it was eradicated in 1980. However, smallpox has emerged as the most threatening bio-terrorism agent; as the first- and second-generation smallpox vaccines have been controversial and have caused severe adverse reactions, new demands for safe smallpox vaccines have been raised and some attenuated smallpox vaccines have been developed. We have developed a cell culture-based highly attenuated third-generation smallpox vaccine candidate KVAC103 strain by 103 serial passages of the Lancy-Vaxina strain derived from the Lister in Vero cells. Several clones were selected, taking into consideration their shape, size, and growth rate in mammalian cells. The clones were then inoculated intracerebrally in suckling mice to test for neurovirulence by observing survival. Protective immune responses in adult mice were examined by measuring the levels of neutralization antibodies and IFN-γ expression. Among several clones, clone 7 was considered the best alternative candidate because there was no mortality in suckling mice against a lethal challenge. In addition, enhanced neutralizing antibodies and T-cell mediated IFN-γ production were observed in clone 7-immunized mice. Clone 7 was named "KVAC103" and was used for the skin toxicity test and full-genome analysis. KVAC103-inoculated rabbits showed reduced skin lesions compared to those inoculated with the Lister strain, Lancy-Vaxina. A whole genome analysis of KVAC103 revealed two major deleted regions that might contribute to the reduced virulence of KVAC103 compared to the Lister strain. Phylogenetic inference supported the close relationship with the Lister strain. Collectively, our data demonstrate that KVAC103 holds promise for use as a third-generation smallpox vaccine strain due to its enhanced safety and efficacy.
Collapse
Affiliation(s)
- Heeji Lim
- Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, CheongJu, Chungbuk 28159, Republic of Korea
| | - Hyun Ju In
- Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, CheongJu, Chungbuk 28159, Republic of Korea
| | - You-Jin Kim
- Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, CheongJu, Chungbuk 28159, Republic of Korea
| | - Sundong Jang
- College of Pharmacy, Chungbuk National University, CheongJu, Chungbuk 28160, Republic of Korea
| | - Yun Ha Lee
- Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, CheongJu, Chungbuk 28159, Republic of Korea
| | - Su Hwan Kim
- Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, CheongJu, Chungbuk 28159, Republic of Korea
| | - Sun Hwa Lee
- Department of Laboratory Medicine, KU Medicine, Seoul 02841, Republic of Korea
| | - Jun Hyuk Park
- Department of Laboratory Medicine Chungcheongnam-do Institute of Health and Environment Research, Hongseong 32254, Republic of Korea
| | - Hyo Jin Yang
- Korea Disease Control and Prevention Agency, CheongJu, Chungbuk 28159, Republic of Korea
| | - Jung-Sik Yoo
- Center for Infectious Disease Research, National Institute of Infectious Diseases, National Institute of Health, CheongJu, Chungbuk 28159, Republic of Korea
| | - Sang-Won Lee
- Korea Disease Control and Prevention Agency, CheongJu, Chungbuk 28159, Republic of Korea
| | - Mi Young Kim
- Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, CheongJu, Chungbuk 28159, Republic of Korea
| | - Gyung Tae Chung
- Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, CheongJu, Chungbuk 28159, Republic of Korea
| | - Sang Gu Yeo
- Division of Infectious Diseases, Sejong Institute of Health and Environment Research, Sejong City 30015, Republic of Korea.
| |
Collapse
|
19
|
Salerno-Gonçalves R, Chen WH, Mulligan MJ, Frey SE, Stapleton JT, Keitel WA, Bailey J, Sendra E, Hill H, Johnson RA, Sztein MB. Vaccine-related major cutaneous reaction size correlates with cellular-mediated immune responses after tularaemia immunisation. Clin Transl Immunology 2021; 10:e1239. [PMID: 33505681 PMCID: PMC7814273 DOI: 10.1002/cti2.1239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 01/31/2023] Open
Abstract
Objectives Francisella tularensis, the causative agent of tularaemia, is an exceptionally infectious bacterium, potentially fatal for humans if left untreated and with the potential to be developed as a bioweapon. Both natural infection and live-attenuated vaccine strain (LVS) confer good protection against tularaemia. LVS vaccination is traditionally administered by scarification, and the formation of a cutaneous reaction or take at the vaccination site is recognised as a clinical correlate of protection. Although previous studies have suggested that high antibody titres following vaccination might serve as a useful surrogate marker, the immunological correlates of protection remain unknown. Methods We investigated the host T-cell-mediated immune (T-CMI) responses elicited following immunisation with LVS vaccine formulated by the DynPort Vaccine Company (DVC-LVS) or the United States Army Medical Research Institute of Infectious Diseases (USAMRIID-LVS). We compared T-CMI responses prompted by these vaccines and correlated them with take size. Results We found that both LVS vaccines elicited similar T-CMI responses. Interestingly, take size associated with the T cells' ability to proliferate, secrete IFN-γ and mobilise degranulation, suggesting that these responses play an essential role in tularaemia protection. Conclusions These results renew the appreciation for vaccination through the scarification as a prime route of inoculation to target pathogens driving specific T-CMI responses and provide further evidence that T-CMI plays a role in protection from tularaemia.
Collapse
Affiliation(s)
- Rosangela Salerno-Gonçalves
- Center for Vaccine Development and Global Health (CVD) University of Maryland School of Medicine Baltimore MD USA
| | - Wilbur H Chen
- Center for Vaccine Development and Global Health (CVD) University of Maryland School of Medicine Baltimore MD USA
| | - Mark J Mulligan
- The Hope Clinic of the Emory Vaccine Center Department of Medicine Division of Infectious Diseases Emory University School of Medicine Decatur GA USA.,Present address: NYU Langone Vaccine Center NYU Grossman School of Medicine Alexandria Center for Life Sciences (West Tower) New York NY USA
| | - Sharon E Frey
- Division of Infectious Diseases, Allergy and Immunology Saint Louis University School of Medicine St. Louis MO USA
| | - Jack T Stapleton
- Iowa City Veterans Administration and the University of Iowa Iowa City IA USA
| | - Wendy A Keitel
- Departments of Molecular Virology and Microbiology, and Medicine Baylor College of Medicine Houston TX USA
| | - Jason Bailey
- Emmes Rockville MD USA.,Present address: Armed Forces Health Surveillance Branch, Integrated Biosurveillance Silver Spring MD USA
| | | | | | - Robert A Johnson
- Biomedical Advanced Research and Development Authority Department of Health and Human Services ASPR Washington DC USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health (CVD) University of Maryland School of Medicine Baltimore MD USA.,Program in Oncology University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center Baltimore MD USA
| |
Collapse
|
20
|
Epicutaneous immunization with modified vaccinia Ankara viral vectors generates superior T cell immunity against a respiratory viral challenge. NPJ Vaccines 2021; 6:1. [PMID: 33398010 PMCID: PMC7782760 DOI: 10.1038/s41541-020-00265-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
Modified Vaccinia Ankara (MVA) was recently approved as a smallpox vaccine. Variola is transmitted by respiratory droplets and MVA immunization by skin scarification (s.s.) protected mice far more effectively against lethal respiratory challenge with vaccinia virus (VACV) than any other route of delivery, and at lower doses. Comparisons of s.s. with intradermal, subcutaneous, or intramuscular routes showed that MVAOVA s.s.-generated T cells were both more abundant and transcriptionally unique. MVAOVA s.s. produced greater numbers of lung Ova-specific CD8+ TRM and was superior in protecting mice against lethal VACVOVA respiratory challenge. Nearly as many lung TRM were generated with MVAOVA s.s. immunization compared to intra-tracheal immunization with MVAOVA and both routes vaccination protected mice against lethal pulmonary challenge with VACVOVA. Strikingly, MVAOVA s.s.-generated effector T cells exhibited overlapping gene transcriptional profiles to those generated via intra-tracheal immunization. Overall, our data suggest that heterologous MVA vectors immunized via s.s. are uniquely well-suited as vaccine vectors for respiratory pathogens, which may be relevant to COVID-19. In addition, MVA delivered via s.s. could represent a more effective dose-sparing smallpox vaccine.
Collapse
|
21
|
Harnessing Cellular Immunity for Vaccination against Respiratory Viruses. Vaccines (Basel) 2020. [DOI: 10.3390/vaccines8040783
expr 839529059 + 832255227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Severe respiratory viral infections, such as influenza, metapneumovirus (HMPV), respiratory syncytial virus (RSV), rhinovirus (RV), and coronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), cause significant mortality and morbidity worldwide. These viruses have been identified as important causative agents of acute respiratory disease in infants, the elderly, and immunocompromised individuals. Clinical signs of infection range from mild upper respiratory illness to more serious lower respiratory illness, including bronchiolitis and pneumonia. Additionally, these illnesses can have long-lasting impact on patient health well beyond resolution of the viral infection. Aside from influenza, there are currently no licensed vaccines against these viruses. However, several research groups have tested various vaccine candidates, including those that utilize attenuated virus, virus-like particles (VLPs), protein subunits, and nanoparticles, as well as recent RNA vaccines, with several of these approaches showing promise. Historically, vaccine candidates have advanced, dependent upon the ability to activate the humoral immune response, specifically leading to strong B cell responses and neutralizing antibody production. More recently, it has been recognized that the cellular immune response is also critical in proper resolution of viral infection and protection against detrimental immunopathology associated with severe disease and therefore, must also be considered when analyzing the efficacy and safety of vaccine candidates. These candidates would ideally result in robust CD4+ and CD8+ T cell responses as well as high-affinity neutralizing antibody. This review will aim to summarize established and new approaches that are being examined to harness the cellular immune response during respiratory viral vaccination.
Collapse
|
22
|
Lukacs NW, Malinczak CA. Harnessing Cellular Immunity for Vaccination against Respiratory Viruses. Vaccines (Basel) 2020; 8:783. [PMID: 33371275 PMCID: PMC7766447 DOI: 10.3390/vaccines8040783] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Severe respiratory viral infections, such as influenza, metapneumovirus (HMPV), respiratory syncytial virus (RSV), rhinovirus (RV), and coronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), cause significant mortality and morbidity worldwide. These viruses have been identified as important causative agents of acute respiratory disease in infants, the elderly, and immunocompromised individuals. Clinical signs of infection range from mild upper respiratory illness to more serious lower respiratory illness, including bronchiolitis and pneumonia. Additionally, these illnesses can have long-lasting impact on patient health well beyond resolution of the viral infection. Aside from influenza, there are currently no licensed vaccines against these viruses. However, several research groups have tested various vaccine candidates, including those that utilize attenuated virus, virus-like particles (VLPs), protein subunits, and nanoparticles, as well as recent RNA vaccines, with several of these approaches showing promise. Historically, vaccine candidates have advanced, dependent upon the ability to activate the humoral immune response, specifically leading to strong B cell responses and neutralizing antibody production. More recently, it has been recognized that the cellular immune response is also critical in proper resolution of viral infection and protection against detrimental immunopathology associated with severe disease and therefore, must also be considered when analyzing the efficacy and safety of vaccine candidates. These candidates would ideally result in robust CD4+ and CD8+ T cell responses as well as high-affinity neutralizing antibody. This review will aim to summarize established and new approaches that are being examined to harness the cellular immune response during respiratory viral vaccination.
Collapse
Affiliation(s)
- Nicholas W. Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA;
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
23
|
Petersen E, Kantele A, Koopmans M, Asogun D, Yinka-Ogunleye A, Ihekweazu C, Zumla A. Human Monkeypox: Epidemiologic and Clinical Characteristics, Diagnosis, and Prevention. Infect Dis Clin North Am 2019; 33:1027-1043. [PMID: 30981594 PMCID: PMC9533922 DOI: 10.1016/j.idc.2019.03.001] [Citation(s) in RCA: 396] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recently, concern has been raised about the emergence of human monkeypox virus and the occasionally severe clinical presentation bearing resemblance to that of smallpox. In 2018 3 patients in the UK were diagnosed with monkeypox, and the frequency and geographic distribution of cases across West and Central Africa have increased in recent years. In Nigeria, most monkeypox patients are aged <40 years and lack cross-protective immunity because they were born after discontinuation of the smallpox eradication campaign. This article reviews the epidemiology, clinical features, and management of monkeypox and discusses its growing public health threat in this context.
Collapse
Affiliation(s)
- Eskild Petersen
- Institute of Clinical Medicine, University of Aarhus, Palle Juul-Jensens Boulevard 82, Aarhus N DK-8200, Denmark; The Royal Hospital, Muscat, Oman; European Society for Clinical Microbiology and Infectious Diseases, Task Force for Emerging Infections, Basel, Switzerland.
| | - Anu Kantele
- Inflammation Center, Helsinki University Hospital and Helsinki University, Stenbäckinkatu 9, PO BOX 100, Helsinki FI-00029 HUS, Finland
| | - Marion Koopmans
- Viroscience Department, Erasmus Medical Centre, Postbus 2040, Rotterdam 3000 CA, the Netherlands
| | - Danny Asogun
- Department of Public Health, College of Medicine, Ambrose Alli University, Ekpoma, Nigeria; Department of Public Health, and Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | | | - Chikwe Ihekweazu
- Nigeria Centre for Disease Control, Plot 801, Ebitu Ukiwe Street, Jabi, Abuja, Nigeria
| | - Alimuddin Zumla
- Division of Infection and Immunity, Center for Clinical Microbiology, University College London, The National Institute of Health Research Biomedical Research Centre at UCL Hospitals, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
24
|
Mucker EM, Wollen-Roberts SE, Kimmel A, Shamblin J, Sampey D, Hooper JW. Intranasal monkeypox marmoset model: Prophylactic antibody treatment provides benefit against severe monkeypox virus disease. PLoS Negl Trop Dis 2018; 12:e0006581. [PMID: 29927927 PMCID: PMC6029809 DOI: 10.1371/journal.pntd.0006581] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 07/03/2018] [Accepted: 06/04/2018] [Indexed: 02/08/2023] Open
Abstract
Concerns regarding outbreaks of human monkeypox or the potential reintroduction of smallpox into an immunological naïve population have prompted the development of animal models and countermeasures. Here we present a marmoset model of monkeypox and smallpox disease utilizing a relevant poxvirus via a natural exposure route. We found that 1000 plaque forming units (PFU) of Monkeypox virus was sufficient to recapitulate smallpox disease, to include an incubation period of approximately 13 days, followed by the onset of rash, and death between 15 and 17 days. Temporally accurate manifestation of viremia and oral shedding were also features. The number of lesions ranged from no lesions to 299, the most reported in a marmoset exposed to a poxvirus. To both evaluate the efficacy of our antibodies and the applicability of the model system, marmosets were prophylactically treated with two monoclonal antibodies, c7D11 and c8A. Of three marmosets, two were completely free of disease and a single marmoset died 8 days after the mock (n = 1) or PBS control(s) (n = 2). Evaluation of the serum levels of the three animals provided a possible explanation to the animal succumbing to disease. Interestingly, more females had lesions (and a greater number of lesions) and lower viral burden (viremia and oral shedding) than males in our studies, suggesting a possible gender effect.
Collapse
Affiliation(s)
- Eric M. Mucker
- United States Army Medical Research Institute of Infectious Diseases, Virology Division, Fort Detrick Maryland, United States of America
| | - Suzanne E. Wollen-Roberts
- United States Army Medical Research Institute of Infectious Diseases, Virology Division, Fort Detrick Maryland, United States of America
| | - Adrienne Kimmel
- United States Army Medical Research Institute of Infectious Diseases, Virology Division, Fort Detrick Maryland, United States of America
| | - Josh Shamblin
- United States Army Medical Research Institute of Infectious Diseases, Virology Division, Fort Detrick Maryland, United States of America
| | - Darryl Sampey
- BioFactura, Inc, Frederick, Maryland, United States of America
| | - Jay W. Hooper
- United States Army Medical Research Institute of Infectious Diseases, Virology Division, Fort Detrick Maryland, United States of America
| |
Collapse
|
25
|
Melamed S, Israely T, Paran N. Challenges and Achievements in Prevention and Treatment of Smallpox. Vaccines (Basel) 2018; 6:vaccines6010008. [PMID: 29382130 PMCID: PMC5874649 DOI: 10.3390/vaccines6010008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/15/2018] [Accepted: 01/26/2018] [Indexed: 01/17/2023] Open
Abstract
Declaration of smallpox eradication by the WHO in 1980 led to discontinuation of the worldwide vaccination campaign. The increasing percentage of unvaccinated individuals, the existence of its causative infectious agent variola virus (VARV), and the recent synthetic achievements increase the threat of intentional or accidental release and reemergence of smallpox. Control of smallpox would require an emergency vaccination campaign, as no other protective measure has been approved to achieve eradication and ensure worldwide protection. Experimental data in surrogate animal models support the assumption, based on anecdotal, uncontrolled historical data, that vaccination up to 4 days postexposure confers effective protection. The long incubation period, and the uncertainty of the exposure status in the surrounding population, call for the development and evaluation of safe and effective methods enabling extension of the therapeutic window, and to reduce the disease manifestations and vaccine adverse reactions. To achieve these goals, we need to evaluate the efficacy of novel and already licensed vaccines as a sole treatment, or in conjunction with immune modulators and antiviral drugs. In this review, we address the available data, recent achievements, and open questions.
Collapse
Affiliation(s)
- Sharon Melamed
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness-Ziona 74100, Israel.
| | - Tomer Israely
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness-Ziona 74100, Israel.
| | - Nir Paran
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness-Ziona 74100, Israel.
| |
Collapse
|
26
|
Development of an animal model of progressive vaccinia in nu/nu mice and the use of bioluminescence imaging for assessment of the efficacy of monoclonal antibodies against vaccinial B5 and L1 proteins. Antiviral Res 2017; 144:8-20. [PMID: 28495463 DOI: 10.1016/j.antiviral.2017.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 11/24/2022]
Abstract
Bioluminescence imaging (BLI) was used to follow dissemination of recombinant vaccinia virus (VACV) expressing luciferase (IHD-J-Luc) in BALB/c nu/nu mice treated post-challenge with monoclonal antibodies (MAbs) against L1 and B5 VACV proteins in a model of Progressive Vaccinia (PV). Areas Under the flux Curve (AUC) were calculated for viral loads in multiple organs in individual mice. Following scarification with 105 pfu, IHD-J-Luc VACV undergoes fast replication at the injection site and disseminates rapidly to the inguinal lymph nodes followed by spleen, liver, and axillary lymph nodes within 2-3 days and before primary lesions are visible at the site of scarification. Extension of survival in nude mice treated with a combination of anti-B5 and anti-L1 MAbs 24 h post challenge correlated with a significant reduction in viral load at the site of scarification and delayed systemic dissemination. Nude mice reconstituted with 104 T cells prior to challenge with IHD-J-Luc, and treated with MAbs post-challenge, survived infection, cleared the virus from all organs and scarification site, and developed anti-VACV IgG and VACV-specific polyfunctional CD8+ T cells that co-expressed the degranulation marker CD107a, and IFNγ and TNFα cytokines. All T cell reconstituted mice survived intranasal re-challenge with IHD-J-Luc (104 pfu) two months after the primary infection. Thus, using BLI to monitor VACV replication in a PV model, we showed that anti-VACV MAbs administered post challenge extended survival of nude mice and protected T cell reconstituted nude mice from lethality by reducing replication at the site of scarification and systemic dissemination of VACV.
Collapse
|
27
|
Gilchuk P, Knight FC, Wilson JT, Joyce S. Eliciting Epitope-Specific CD8+ T Cell Response by Immunization with Microbial Protein Antigens Formulated with α-Galactosylceramide: Theory, Practice, and Protocols. Methods Mol Biol 2017; 1494:321-352. [PMID: 27718206 DOI: 10.1007/978-1-4939-6445-1_25] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD8+ cytotoxic T lymphocytes confer protection against infectious diseases caused by viruses, bacteria, and parasites. Hence, significant efforts have been invested into devising ways to generate CD8+ T cell-targeted vaccines. Generation of microbe-free protein subunit vaccines requires a thorough knowledge of protective target antigens. Such antigens are proteolytically processed peptides presented by MHC class I molecules. To induce a robust antigen-specific CD8+ T cell response through vaccination, it is essential to formulate the antigen with an effective adjuvant. Here, we describe a versatile method for generating high-frequency antigen-specific CD8+ T cells through immunization of mice using the invariant natural killer T cell agonist α-galactosylceramide as the adjuvant.
Collapse
Affiliation(s)
- Pavlo Gilchuk
- Veterans Administration Tennessee Valley Healthcare System, US Department of Veterans Affairs, Nashville, TN, 37332, USA.,Department of Pathology, Microbiology and Immunology, School of Medicine, Vanderbilt University, A4223 Medical Centre North, 1161 21st Avenue South, Nashville, TN, 37332, USA
| | - Frances C Knight
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, TN, 37332, USA
| | - John T Wilson
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, TN, 37332, USA.,Department of Chemical & Biomolecular Engineering, School of Engineering, Vanderbilt University, Nashville, TN, 37332, USA
| | - Sebastian Joyce
- Veterans Administration Tennessee Valley Healthcare System, US Department of Veterans Affairs, Nashville, TN, 37332, USA. .,Department of Pathology, Microbiology and Immunology, School of Medicine, Vanderbilt University, A4223 Medical Centre North, 1161 21st Avenue South, Nashville, TN, 37332, USA.
| |
Collapse
|
28
|
Kennedy RB, Poland GA, Ovsyannikova IG, Oberg AL, Asmann YW, Grill DE, Vierkant RA, Jacobson RM. Impaired innate, humoral, and cellular immunity despite a take in smallpox vaccine recipients. Vaccine 2016; 34:3283-90. [PMID: 27177944 PMCID: PMC5528000 DOI: 10.1016/j.vaccine.2016.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 11/29/2022]
Abstract
Smallpox vaccine is highly effective, inducing protective immunity to smallpox and diseases caused by related orthopoxviruses. Smallpox vaccine efficacy was historically defined by the appearance of a lesion or "take" at the vaccine site, which leaves behind a characteristic scar. Both the take and scar are readily recognizable and were used during the eradication effort to indicate successful vaccination and to categorize individuals as "protected." However, the development of a typical vaccine take may not equate to the successful development of a robust, protective immune response. In this report, we examined two large (>1000) cohorts of recipients of either Dryvax(®) or ACAM2000 using a testing and replication study design and identified subgroups of individuals who had documented vaccine takes, but who failed to develop robust neutralizing antibody titers. Examination of these individuals revealed that they had suboptimal cellular immune responses as well. Further testing indicated these low responders had a diminished innate antiviral gene expression pattern (IFNA1, CXCL10, CXCL11, OASL) upon in vitro stimulation with vaccinia virus, perhaps indicative of a dysregulated innate response. Our results suggest that poor activation of innate antiviral pathways may result in suboptimal immune responses to the smallpox vaccine. These genes and pathways may serve as suitable targets for adjuvants in new attenuated smallpox vaccines and/or effective antiviral therapy targets against poxvirus infections.
Collapse
Affiliation(s)
- Richard B Kennedy
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, MN, USA; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Gregory A Poland
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, MN, USA; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Inna G Ovsyannikova
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, MN, USA; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ann L Oberg
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Yan W Asmann
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Diane E Grill
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Robert A Vierkant
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Robert M Jacobson
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, MN, USA; Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA; Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
29
|
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
|
30
|
Eto A, Saito T, Yokote H, Kurane I, Kanatani Y. Recent advances in the study of live attenuated cell-cultured smallpox vaccine LC16m8. Vaccine 2015; 33:6106-11. [PMID: 26319072 PMCID: PMC9533910 DOI: 10.1016/j.vaccine.2015.07.111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/20/2015] [Accepted: 07/29/2015] [Indexed: 12/02/2022]
Abstract
LC16m8 is a live, attenuated, cell-cultured smallpox vaccine that was developed and licensed in Japan in the 1970s, but was not used in the campaign to eradicate smallpox. In the early 2000s, the potential threat of bioterrorism led to reconsideration of the need for a smallpox vaccine. Subsequently, LC16m8 production was restarted in Japan in 2002, requiring re-evaluation of its safety and efficacy. Approximately 50,000 children in the 1970s and about 3500 healthy adults in the 2000s were vaccinated with LC16m8 in Japan, and 153 adults have been vaccinated with LC16m8 or Dryvax in phase I/II clinical trials in the USA. These studies confirmed the safety and efficacy of LC16m8, while several studies in animal models have shown that LC16m8 protects the host against viral challenge. The World Health Organization Strategic Advisory Group of Experts on Immunization recommended LC16m8, together with ACAM2000, as a stockpile vaccine in 2013. In addition, LC16m8 is expected to be a viable alternative to first-generation smallpox vaccines to prevent human monkeypox.
Collapse
Affiliation(s)
- Akiko Eto
- Department of Health Crisis Management, National Institute of Public Health, 2-3-6 Minami, Wako-shi, 351-0197, Saitama, Japan
| | - Tomoya Saito
- Department of Health Crisis Management, National Institute of Public Health, 2-3-6 Minami, Wako-shi, 351-0197, Saitama, Japan
| | - Hiroyuki Yokote
- Chemo-Sero-Therapeutic Research Institute (Kaketsuken), 1-6-1 Okubo, Kita-ku, Kumamoto-shi, 860-8568, Kumamoto, Japan
| | - Ichiro Kurane
- National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, 162-8640, Tokyo, Japan
| | - Yasuhiro Kanatani
- Department of Health Crisis Management, National Institute of Public Health, 2-3-6 Minami, Wako-shi, 351-0197, Saitama, Japan.
| |
Collapse
|
31
|
Nishiyama Y, Fujii T, Kanatani Y, Shinmura Y, Yokote H, Hashizume S. Freeze-dried live attenuated smallpox vaccine prepared in cell culture "LC16-KAKETSUKEN": Post-marketing surveillance study on safety and efficacy compliant with Good Clinical Practice. Vaccine 2015; 33:6120-7. [PMID: 26455406 DOI: 10.1016/j.vaccine.2015.09.067] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 08/28/2015] [Accepted: 09/18/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND In Japan, production of smallpox vaccine LC16m8 (named LC16-KAKETSUKEN) was restarted and was determined to be maintained as a national stockpile in March 2002. OBJECTIVE To conduct a post-marketing surveillance study of the vaccination of freeze-dried live attenuated smallpox vaccine prepared in cell culture LC16-KAKETSUKEN using attenuated vaccinia strain LC16m8. The study complied with Good Clinical Practice, focusing on a comparison between primary vaccinees and re-vaccinees. METHOD 268 personnel (261 males and 7 females) of the Japan Ground Self-Defense Force were inoculated with LC16-KAKETSUKEN and thereafter adverse events and efficacy were evaluated. RESULTS Among 268 vaccinee participants, the following vaccinees showed adverse events, none serious: 53 of 196 primary vaccinees (without previous smallpox vaccination), 4 of 71 re-vaccinees (with previous smallpox vaccination) and 1 vaccinee with unknown previous vaccination history. A breakdown of adverse events observed in this study (total 268 vaccinees) showed the following minor or mild adverse events: 52 (19.4%) swelling of axillary lymph node, 4 (1.5%) fever, 2 (0.7%) fatigue, 1 (0.4%) of rash, 14 (5.2%) erythema at the inoculation site, 1 (0.4%) swelling at the inoculation site and 1 (0.4%) autoinoculation. The incidence of adverse events for primary vaccinees (53/196; 27.0%) was significantly higher than for re-vaccinees (4/71; 5.6%). However, the proportion of vaccine take was significantly higher for primary vaccinees (185/196; 94.4%) than for re-vaccinees (58/71; 81.7%). Although the proportion of vaccine take of re-vaccinees was significantly lower than for primary vaccinees due to preexisting immunity by previous vaccination, no significant difference was found in neutralizing antibody titers between primary vaccinees and re-vaccinees at 1, 4 and 7 months after LC16-KAKETSUKEN vaccination. CONCLUSION The present post-marketing surveillance study compliant with Good Clinical Practice demonstrated the efficacy and safety of the smallpox vaccine LC16-KAKETSUKEN in an adult population. LC16-KAKETSUKEN is the sole currently available licensed smallpox vaccine for both adult and pediatric populations.
Collapse
Affiliation(s)
- Yasumasa Nishiyama
- Health Care Center, Japan Self-Defense Forces Central Hospital, 1-2-24 Ikeziri, Setagaya-ku, Tokyo 154-8532, Japan
| | - Tatsuya Fujii
- Health Care Center, Japan Self-Defense Forces Central Hospital, 1-2-24 Ikeziri, Setagaya-ku, Tokyo 154-8532, Japan
| | - Yasuhiro Kanatani
- National Institute of Public Health, 2-3-6 Minami, Wako-shi, Saitama 351-0197, Japan
| | - Yasuhiko Shinmura
- The Chemo-Sero-Therapeutic Research Institute (Kaketsuken), 1-6-1 Okubo, Kita-ku, Kumamoto-shi, Kumamoto 860-8568, Japan.
| | - Hiroyuki Yokote
- The Chemo-Sero-Therapeutic Research Institute (Kaketsuken), 1-6-1 Okubo, Kita-ku, Kumamoto-shi, Kumamoto 860-8568, Japan
| | - So Hashizume
- Chiba University, 1-33, Yayoicho, Inage Ward, Chiba-shi, Chiba 263-8522, Japan
| |
Collapse
|
32
|
Genomic Analysis, Phenotype, and Virulence of the Historical Brazilian Smallpox Vaccine Strain IOC: Implications for the Origins and Evolutionary Relationships of Vaccinia Virus. J Virol 2015; 89:11909-25. [PMID: 26378174 DOI: 10.1128/jvi.01833-15] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Smallpox was declared eradicated in 1980 after an intensive vaccination program using different strains of vaccinia virus (VACV; Poxviridae). VACV strain IOC (VACV-IOC) was the seed strain of the smallpox vaccine manufactured by the major vaccine producer in Brazil during the smallpox eradication program. However, little is known about the biological and immunological features as well as the phylogenetic relationships of this first-generation vaccine. In this work, we present a comprehensive characterization of two clones of VACV-IOC. Both clones had low virulence in infected mice and induced a protective immune response against a lethal infection comparable to the response of the licensed vaccine ACAM2000 and the parental strain VACV-IOC. Full-genome sequencing revealed the presence of several fragmented virulence genes that probably are nonfunctional, e.g., F1L, B13R, C10L, K3L, and C3L. Most notably, phylogenetic inference supported by the structural analysis of the genome ends provides evidence of a novel, independent cluster in VACV phylogeny formed by VACV-IOC, the Brazilian field strains Cantagalo (CTGV) and Serro 2 viruses, and horsepox virus, a VACV-like virus supposedly related to an ancestor of the VACV lineage. Our data strongly support the hypothesis that CTGV-like viruses represent feral VACV that evolved in parallel with VACV-IOC after splitting from a most recent common ancestor, probably an ancient smallpox vaccine strain related to horsepox virus. Our data, together with an interesting historical investigation, revisit the origins of VACV and propose new evolutionary relationships between ancient and extant VACV strains, mainly horsepox virus, VACV-IOC/CTGV-like viruses, and Dryvax strain. IMPORTANCE First-generation vaccines used to eradicate smallpox had rates of adverse effects that are not acceptable by current health care standards. Moreover, these vaccines are genetically heterogeneous and consist of a pool of quasispecies of VACV. Therefore, the search for new-generation smallpox vaccines that combine low pathogenicity, immune protection, and genetic homogeneity is extremely important. In addition, the phylogenetic relationships and origins of VACV strains are quite nebulous. We show the characterization of two clones of VACV-IOC, a unique smallpox vaccine strain that contributed to smallpox eradication in Brazil. The immunogenicity and reduced virulence make the IOC clones good options for alternative second-generation smallpox vaccines. More importantly, this study reveals the phylogenetic relationship between VACV-IOC, feral VACV established in nature, and the ancestor-like horsepox virus. Our data expand the discussion on the origins and evolutionary connections of VACV lineages.
Collapse
|
33
|
Reply to "Use of the LC16m8 Smallpox Vaccine in Immunocompromised Individuals Is Still Too Risky". CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:605. [PMID: 25921467 DOI: 10.1128/cvi.00055-15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
34
|
Vaccinia virus strain LC16m8 defective in the B5R gene keeps strong protection comparable to its parental strain Lister in immunodeficient mice. Vaccine 2015; 33:6112-9. [PMID: 26241947 DOI: 10.1016/j.vaccine.2015.07.076] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/19/2015] [Accepted: 07/20/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Attenuated vaccinia virus strain, LC16m8, defective in the B5R envelope protein gene, is used as a stockpile smallpox vaccine strain in Japan against bioterrorism: the defect in the B5R gene mainly contributes to its highly attenuated properties. METHODS The protective activity of LC16m8 vaccine against challenge with a lethal dose of vaccinia Western Reserve strain was assessed in wild-type and immunodeficient mice lacking CD4, MHC class I, MHC class II or MHC class I and II antigens. RESULTS The immunization with LC16m8 induced strong protective activity comparable to that of its parent strain, Lister (Elstree) strain, in wild-type mice from 2 days to 1 year after vaccination, as well as in immunodeficient mice at 2 or 3 weeks after vaccination. These results implicated that the defect in the B5R gene hardly affected the potential activity of LC16m8 to induce innate, cell-mediated and humoral immunity, and that LC16m8 could be effective in immunodeficient patients. CONCLUSION LC16m8 with truncated B5 protein has an activity to induce immunity, such as innate immunity and subsequent cell-mediated and humoral immunity almost completely comparable to the activity of its parental strain Lister.
Collapse
|
35
|
Medcalf S, Bilek L, Hartman T, Iwen PC, Leuschen P, Miller H, O'Keefe A, Sayles H, Smith PW. Smallpox Vaccination of Laboratory Workers at US Variola Testing Sites. Emerg Infect Dis 2015. [PMID: 26196153 PMCID: PMC4517736 DOI: 10.3201/eid2108.140956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
To evaluate the need to revaccinate laboratory workers against smallpox, we assessed regular revaccination at the US Laboratory Response Network’s variola testing sites by examining barriers to revaccination and the potential for persistence of immunity. Our data do not provide evidence to suggest prolonging the recommended interval for revaccination.
Collapse
|
36
|
Flesch IEA, Hollett NA, Wong YC, Quinan BR, Howard D, da Fonseca FG, Tscharke DC. Extent of Systemic Spread Determines CD8+ T Cell Immunodominance for Laboratory Strains, Smallpox Vaccines, and Zoonotic Isolates of Vaccinia Virus. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26195812 DOI: 10.4049/jimmunol.1402508] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
CD8(+) T cells that recognize virus-derived peptides presented on MHC class I are vital antiviral effectors. Such peptides presented by any given virus vary greatly in immunogenicity, allowing them to be ranked in an immunodominance hierarchy. However, the full range of parameters that determine immunodominance and the underlying mechanisms remain unknown. In this study, we show across a range of vaccinia virus strains, including the current clonal smallpox vaccine, that the ability of a strain to spread systemically correlated with reduced immunodominance. Reduction in immunodominance was observed both in the lymphoid system and at the primary site of infection. Mechanistically, reduced immunodominance was associated with more robust priming and especially priming in the spleen. Finally, we show this is not just a property of vaccine and laboratory strains of virus, because an association between virulence and immunodominance was also observed in isolates from an outbreak of zoonotic vaccinia virus that occurred in Brazil.
Collapse
Affiliation(s)
- Inge E A Flesch
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Natasha A Hollett
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Yik Chun Wong
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Bárbara Resende Quinan
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 2601, Australia; Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; and
| | - Debbie Howard
- John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Flávio G da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; and
| | - David C Tscharke
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory 2601, Australia; John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| |
Collapse
|
37
|
Sánchez-Sampedro L, Perdiguero B, Mejías-Pérez E, García-Arriaza J, Di Pilato M, Esteban M. The evolution of poxvirus vaccines. Viruses 2015; 7:1726-803. [PMID: 25853483 PMCID: PMC4411676 DOI: 10.3390/v7041726] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/16/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023] Open
Abstract
After Edward Jenner established human vaccination over 200 years ago, attenuated poxviruses became key players to contain the deadliest virus of its own family: Variola virus (VARV), the causative agent of smallpox. Cowpox virus (CPXV) and horsepox virus (HSPV) were extensively used to this end, passaged in cattle and humans until the appearance of vaccinia virus (VACV), which was used in the final campaigns aimed to eradicate the disease, an endeavor that was accomplished by the World Health Organization (WHO) in 1980. Ever since, naturally evolved strains used for vaccination were introduced into research laboratories where VACV and other poxviruses with improved safety profiles were generated. Recombinant DNA technology along with the DNA genome features of this virus family allowed the generation of vaccines against heterologous diseases, and the specific insertion and deletion of poxvirus genes generated an even broader spectrum of modified viruses with new properties that increase their immunogenicity and safety profile as vaccine vectors. In this review, we highlight the evolution of poxvirus vaccines, from first generation to the current status, pointing out how different vaccines have emerged and approaches that are being followed up in the development of more rational vaccines against a wide range of diseases.
Collapse
MESH Headings
- Animals
- History, 18th Century
- History, 19th Century
- History, 20th Century
- History, 21st Century
- Humans
- Poxviridae/immunology
- Poxviridae/isolation & purification
- Smallpox/prevention & control
- Smallpox Vaccine/history
- Smallpox Vaccine/immunology
- Smallpox Vaccine/isolation & purification
- Vaccines, Attenuated/history
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/isolation & purification
- Vaccines, Synthetic/history
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/isolation & purification
Collapse
Affiliation(s)
- Lucas Sánchez-Sampedro
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Ernesto Mejías-Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain
| | - Mauro Di Pilato
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| |
Collapse
|
38
|
Hickman HD, Reynoso GV, Ngudiankama BF, Cush SS, Gibbs J, Bennink JR, Yewdell JW. CXCR3 chemokine receptor enables local CD8(+) T cell migration for the destruction of virus-infected cells. Immunity 2015; 42:524-37. [PMID: 25769612 DOI: 10.1016/j.immuni.2015.02.009] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 01/08/2015] [Accepted: 02/20/2015] [Indexed: 12/16/2022]
Abstract
CD8(+) T cells play a critical role in limiting peripheral virus replication, yet how they locate virus-infected cells within tissues is unknown. Here, we have examined the environmental signals that CD8(+) T cells use to localize and eliminate virus-infected skin cells. Epicutaneous vaccinia virus (VV) infection, mimicking human smallpox vaccination, greatly increased expression of the CXCR3 chemokine receptor ligands CXCL9 and CXCL10 in VV-infected skin. Despite normal T cell numbers in the skin, Cxcr3(-/-) mice exhibited dramatically impaired CD8(+)-T-cell-dependent virus clearance. Intravital microscopy revealed that Cxcr3(-/-) T cells were markedly deficient in locating, engaging, and killing virus-infected cells. Further, transfer of wild-type CD8(+) T cells restored viral clearance in Cxcr3(-/-) animals. These findings demonstrate a function for CXCR3 in enhancing the ability of tissue-localized CD8(+) T cells to locate virus-infected cells and thereby exert anti-viral effector functions.
Collapse
Affiliation(s)
- Heather D Hickman
- Cell Biology and Viral Immunology Sections, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA.
| | - Glennys V Reynoso
- Cell Biology and Viral Immunology Sections, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Barbara F Ngudiankama
- Cell Biology and Viral Immunology Sections, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Stephanie S Cush
- Cell Biology and Viral Immunology Sections, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - James Gibbs
- Cell Biology and Viral Immunology Sections, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Jack R Bennink
- Cell Biology and Viral Immunology Sections, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Jonathan W Yewdell
- Cell Biology and Viral Immunology Sections, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
39
|
Burwitz BJ, Reed JS, Hammond KB, Ohme MA, Planer SL, Legasse AW, Ericsen AJ, Richter Y, Golomb G, Sacha JB. Technical advance: liposomal alendronate depletes monocytes and macrophages in the nonhuman primate model of human disease. J Leukoc Biol 2014; 96:491-501. [PMID: 24823811 PMCID: PMC4632165 DOI: 10.1189/jlb.5ta0713-373r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 04/02/2014] [Accepted: 04/22/2014] [Indexed: 01/02/2023] Open
Abstract
Nonhuman primates are critical animal models for the study of human disorders and disease and offer a platform to assess the role of immune cells in pathogenesis via depletion of specific cellular subsets. However, this model is currently hindered by the lack of reagents that safely and specifically ablate myeloid cells of the monocyte/macrophage Lin. Given the central importance of macrophages in homeostasis and host immunity, development of a macrophage-depletion technique in nonhuman primates would open new avenues of research. Here, using LA at i.v. doses as low as 0.1 mg/kg, we show a >50% transient depletion of circulating monocytes and tissue-resident macrophages in RMs by an 11-color flow cytometric analysis. Diminution of monocytes was followed rapidly by emigration of monocytes from the bone marrow, leading to a rebound of monocytes to baseline levels. Importantly, LA was well-tolerated, as no adverse effects or changes in gross organ function were observed during depletion. These results advance the ex vivo study of myeloid cells by flow cytometry and pave the way for in vivo studies of monocyte/macrophage biology in nonhuman primate models of human disease.
Collapse
Affiliation(s)
- Benjamin J Burwitz
- Vaccine and Gene Therapy Institute and Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Jason S Reed
- Vaccine and Gene Therapy Institute and Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Katherine B Hammond
- Vaccine and Gene Therapy Institute and Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Merete A Ohme
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Shannon L Planer
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Alfred W Legasse
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Adam J Ericsen
- Department of Pathology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Gershon Golomb
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Jonah B Sacha
- Vaccine and Gene Therapy Institute and Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA;
| |
Collapse
|
40
|
Safety of attenuated smallpox vaccine LC16m8 in immunodeficient mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:1261-6. [PMID: 24990910 DOI: 10.1128/cvi.00199-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Freeze-dried live attenuated smallpox vaccine LC16m8 prepared in cell culture has been the sole smallpox vaccine licensed in Japan since 1975 and was recently recommended as a WHO stockpile vaccine. We evaluated the safety of recently remanufactured lots of LC16m8 using a series of immunodeficient mouse models. These models included suckling mice, severe combined immunodeficiency disease (SCID) mice, and wild-type mice treated with cyclosporine. LC16m8 showed extremely low virulence in each of the three mouse models compared with that of its parental strains, Lister and LC16mO. These results provide further evidence that LC16m8 is one of the safest replication-competent smallpox vaccines in the world and may be considered for use in immunodeficient patients.
Collapse
|
41
|
Immunogenicity and safety of the vaccinia virus LC16m8Δ vector expressing SIV Gag under a strong or moderate promoter in a recombinant BCG prime-recombinant vaccinia virus boost protocol. Vaccine 2013; 31:3549-57. [DOI: 10.1016/j.vaccine.2013.05.071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 03/23/2013] [Accepted: 05/17/2013] [Indexed: 01/13/2023]
|
42
|
Gilchuk P, Spencer CT, Conant SB, Hill T, Gray JJ, Niu X, Zheng M, Erickson JJ, Boyd KL, McAfee KJ, Oseroff C, Hadrup SR, Bennink JR, Hildebrand W, Edwards KM, Crowe JE, Williams JV, Buus S, Sette A, Schumacher TNM, Link AJ, Joyce S. Discovering naturally processed antigenic determinants that confer protective T cell immunity. J Clin Invest 2013; 123:1976-87. [PMID: 23543059 DOI: 10.1172/jci67388] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 02/07/2013] [Indexed: 12/15/2022] Open
Abstract
CD8+ T cells (TCD8) confer protective immunity against many infectious diseases, suggesting that microbial TCD8 determinants are promising vaccine targets. Nevertheless, current T cell antigen identification approaches do not discern which epitopes drive protective immunity during active infection - information that is critical for the rational design of TCD8-targeted vaccines. We employed a proteomics-based approach for large-scale discovery of naturally processed determinants derived from a complex pathogen, vaccinia virus (VACV), that are presented by the most frequent representatives of four major HLA class I supertypes. Immunologic characterization revealed that many previously unidentified VACV determinants were recognized by smallpox-vaccinated human peripheral blood cells in a variegated manner. Many such determinants were recognized by HLA class I-transgenic mouse immune TCD8 too and elicited protective TCD8 immunity against lethal intranasal VACV infection. Notably, efficient processing and stable presentation of immune determinants as well as the availability of naive TCD8 precursors were sufficient to drive a multifunctional, protective TCD8 response. Our approach uses fundamental insights into T cell epitope processing and presentation to define targets of protective TCD8 immunity within human pathogens that have complex proteomes, suggesting that this approach has general applicability in vaccine sciences.
Collapse
Affiliation(s)
- Pavlo Gilchuk
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Clinical development of Modified Vaccinia virus Ankara vaccines. Vaccine 2013; 31:4241-6. [PMID: 23523410 DOI: 10.1016/j.vaccine.2013.03.020] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 03/11/2013] [Indexed: 12/21/2022]
Abstract
The smallpox vaccine Vaccinia was successfully used to eradicate smallpox, but although very effective, it was a very reactogenic vaccine and responsible for the deaths of one or two people per million vaccinated. Modified Vaccinia virus Ankara (MVA) is a replication-deficient and attenuated derivative, also used in the smallpox eradication campaign and now being developed as a recombinant viral vector to produce vaccines against infectious diseases and cancer. Many clinical trials of these new vaccines have been conducted, and the findings of these trials are reviewed here. The safety of MVA is now well documented, immunogenicity is influenced by the dose and vaccination regimen, and information on the efficacy of MVA-vectored vaccines is now beginning to accumulate.
Collapse
|
44
|
Zhang X, Sobue T, Isshiki M, Makino SI, Inoue M, Kato K, Shioda T, Ohashi T, Sato H, Komano J, Hanabusa H, Shida H. Elicitation of both anti HIV-1 Env humoral and cellular immunities by replicating vaccinia prime Sendai virus boost regimen and boosting by CD40Lm. PLoS One 2012; 7:e51633. [PMID: 23236521 PMCID: PMC3517520 DOI: 10.1371/journal.pone.0051633] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 11/08/2012] [Indexed: 11/18/2022] Open
Abstract
For protection from HIV-1 infection, a vaccine should elicit both humoral and cell-mediated immune responses. A novel vaccine regimen and adjuvant that induce high levels of HIV-1 Env-specific T cell and antibody (Ab) responses was developed in this study. The prime-boost regimen that used combinations of replication-competent vaccinia LC16m8Δ (m8Δ) and Sendai virus (SeV) vectors expressing HIV-1 Env efficiently produced both Env-specific CD8+ T cells and anti-Env antibodies, including neutralizing antibodies (nAbs). These results sharply contrast with vaccine regimens that prime with an Env expressing plasmid and boost with the m8Δ or SeV vector that mainly elicited cellular immunities. Moreover, co-priming with combinations of m8Δs expressing Env or a membrane-bound human CD40 ligand mutant (CD40Lm) enhanced Env-specific CD8+ T cell production, but not anti-Env antibody production. In contrast, priming with an m8Δ that coexpresses CD40Lm and Env elicited more anti-Env Abs with higher avidity, but did not promote T cell responses. These results suggest that the m8Δ prime/SeV boost regimen in conjunction with CD40Lm expression could be used as an immunization platform for driving both potent cellular and humoral immunities against pathogens such as HIV-1.
Collapse
Affiliation(s)
- Xianfeng Zhang
- Institute for Genetic Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Tomoyoshi Sobue
- Institute for Genetic Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Mao Isshiki
- Institute for Genetic Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Shun-ichi Makino
- Institute for Genetic Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Makoto Inoue
- DNAVEC Corporation, Techno Park Oho, Tsukuba, Ibaraki, Japan
| | - Kazunori Kato
- Department of BioMedical Engineering, Toyo University, Kawagoe, Saitama, Japan
| | - Tatsuo Shioda
- Department of Viral Infection, Research Institute for Microbial Disease, Osaka University, Yamada-oka, Suita-shi, Osaka, Japan
| | - Takashi Ohashi
- Institute for Genetic Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Hirotaka Sato
- Institute for Genetic Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Jun Komano
- Division of Virology, Department of Infectious Diseases, Osaka Prefectural Institute of Public Health, Nakamichi Higashinari-ku, Osaka, Japan
| | | | - Hisatoshi Shida
- Institute for Genetic Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
- * E-mail:
| |
Collapse
|
45
|
Kennedy JS, Gurwith M, Yokote H, Greenberg RN. Reply to Friedman. J Infect Dis 2012. [DOI: 10.1093/infdis/jis454] [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] Open
|
46
|
Abstract
In the twentieth century vaccine development has moved from the use of attenuated or killed micro-organisms to protein sub-unit vaccines, with vaccine immunogenicity assessed by measuring antibodies induced by vaccination. However, for many infectious diseases T cells are an important part of naturally acquired protective immune responses, and inducing these by vaccination has been the aim of much research. The progress that has been made in developing effective T-cell-inducing vaccines against viral and parasitic diseases such as HIV and malaria is discussed, along with recent developments in therapeutic vaccine development for chronic viral infections and cancer. Although many ways of inducing T cells by vaccination have been assessed, the majority result in low level, non-protective responses. Sufficient clinical research has now been conducted to establish that replication-deficient viral vectored vaccines lead the field in inducing strong and broad responses, and efficacy studies of T-cell-inducing vaccines against a number of diseases are finally demonstrating that this is a valid approach to filling the gaps in our defence against not only infectious disease, but some forms of cancer.
Collapse
|
47
|
Reynolds MG, Damon IK. Outbreaks of human monkeypox after cessation of smallpox vaccination. Trends Microbiol 2012; 20:80-7. [PMID: 22239910 DOI: 10.1016/j.tim.2011.12.001] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 12/02/2011] [Accepted: 12/08/2011] [Indexed: 11/22/2022]
Abstract
The recent observation of a surge in human monkeypox in the Democratic Republic of the Congo (DRC) prompts the question of whether cessation of smallpox vaccination is driving the phenomenon, and if so, why is re-emergence not universal throughout the historic geographic range of the virus? Research addressing the virus's mechanisms for immune evasion and induction, as well as that directed at elucidating the genes involved in pathogenesis in different viral lineages (West African vs Congo Basin), provide insights to help explain why emergence appears to be geographically limited. Novel vaccines offer one solution to curtail the spread of this disease.
Collapse
Affiliation(s)
- Mary G Reynolds
- Division of High-consequence Pathogens and Pathology, US Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Mailstop G-43, Atlanta, GA 30333, USA
| | | |
Collapse
|
48
|
Denzler KL, Babas T, Rippeon A, Huynh T, Fukushima N, Rhodes L, Silvera PM, Jacobs BL. Attenuated NYCBH vaccinia virus deleted for the E3L gene confers partial protection against lethal monkeypox virus disease in cynomolgus macaques. Vaccine 2011; 29:9684-90. [PMID: 22001879 PMCID: PMC5001690 DOI: 10.1016/j.vaccine.2011.09.135] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2011] [Revised: 09/09/2011] [Accepted: 09/30/2011] [Indexed: 11/16/2022]
Abstract
The New York City Board of Health (NYCBH) vaccinia virus is the currently licensed vaccine for use in the US against smallpox. The vaccine under investigation in this study has been attenuated by deletion of the innate immune evasion gene, E3L, and shown to be protective in homologous virus mouse challenge and heterologous virus mouse and rabbit challenge models. In this study we compared NYCBH deleted for the E3L gene (NYCBHΔE3L) to NYCBH for the ability to induce phosphorylation of proinflammatory signaling proteins and the ability to protect cynomolgus macaques from heterologous challenge with monkeypox virus (MPXV). NYCBHΔE3L induced phosphorylation of PKR and eIF2α as well as p38, SAPK/JNK, and IRF3 which can lead to induction of proinflammatory gene transcription. Vaccination of macaques with two doses of NYCBHΔE3L resulted in negligible pock formation at the site of scarification in comparison to vaccination using a single dose of NYCBH, but still elicited neutralizing antibodies and protected 75% of the animals from mortality after challenge with MPXV. However, NYCBHΔE3L-vaccinated animals developed a high number of secondary skin lesions and blood viral load similar to that seen in unvaccinated controls. The NYCBHΔE3L-vaccinated animals that survived MPXV challenge were able to show resolution of blood viral load, a decrease in number of skin lesions, and an improved clinical score by three weeks post challenge. These results suggest that although the highly attenuated NYCBHΔE3L allows proinflammatory signal transduction to occur, it does not provide full protection against monkeypox challenge.
Collapse
Affiliation(s)
- Karen L Denzler
- Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kennedy JS, Gurwith M, Dekker CL, Frey SE, Edwards KM, Kenner J, Lock M, Empig C, Morikawa S, Saijo M, Yokote H, Karem K, Damon I, Perlroth M, Greenberg RN. Safety and immunogenicity of LC16m8, an attenuated smallpox vaccine in vaccinia-naive adults. J Infect Dis 2011; 204:1395-402. [PMID: 21921208 PMCID: PMC3218648 DOI: 10.1093/infdis/jir527] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 06/07/2011] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION LC16m8 is an attenuated cell culture-adapted Lister vaccinia smallpox vaccine missing the B5R protein and licensed for use in Japan. METHODS We conducted a phase I/II clinical trial that compared the safety and immunogenicity of LC16m8 with Dryvax in vaccinia-naive participants. Adverse events were assessed, as were electrocardiography and laboratory testing for cardiotoxicity and viral culturing of the vaccination sites. Neutralization titers to vaccinia, monkeypox, and variola major were assessed and cell-mediated immune responses were measured by interferon (IFN)-γ enzyme-linked immunosorbent spot and lymphoproliferation assays. RESULTS Local and systemic reactions after vaccination with LC16m8 were similar to those reported after Dryvax. No clinically significant abnormalities consistent with cardiac toxicity were seen for either vaccine. Both vaccines achieved antivaccinia, antivariola, and antimonkeypox neutralizing antibody titers >1:40, although the mean plaque reduction neutralization titer of LC16m8 at day 30 after vaccination was significantly lower than Dryvax for anti-NYCBH vaccinia (P < .01), antimonkeypox (P < .001), and antivariola (P < .001). LC16m8 produced robust cellular immune responses that trended higher than Dryvax for lymphoproliferation (P = .06), but lower for IFN-γ ELISPOT (P = .02). CONCLUSIONS LC16m8 generates neutralizing antibody titers to multiple poxviruses, including vaccinia, monkeypox, and variola major, and broad T-cell responses, indicating that LC16m8 may have efficacy in protecting individuals from smallpox. Clinical Trials Registration. NCT00103584.
Collapse
Affiliation(s)
| | | | - Cornelia L. Dekker
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Stanford University School of Medicine, California
| | - Sharon E. Frey
- Department of Internal Medicine, Division of Infectious Diseases and Immunology, Saint Louis University Health Sciences Center, Missouri
| | - Kathryn M. Edwards
- Department of Pediatrics, Division of Infectious Diseases, Vanderbilt Vaccine Research Program, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Michael Lock
- Statistical Consultant, Mountain View, California
| | - Cyril Empig
- Peregrine Pharmaceuticals, Inc, Tustin, California
| | | | | | - Hiroyuki Yokote
- Chemo-Sero-Therapeutic Research Institute (Kaketsuken), Kumamoto, Japan
| | - Kevin Karem
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Inger Damon
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Mark Perlroth
- Department of Internal Medicine, Stanford University School of Medicine, California
| | - Richard N. Greenberg
- Department of Internal Medicine, University of Kentucky School of Medicine, Lexington
| |
Collapse
|
50
|
Johnson BF, Kanatani Y, Fujii T, Saito T, Yokote H, Smith GL. Serological responses in humans to the smallpox vaccine LC16m8. J Gen Virol 2011; 92:2405-2410. [PMID: 21715598 PMCID: PMC3347799 DOI: 10.1099/vir.0.034207-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In response to potential bioterrorism with smallpox, members of the Japanese Self-Defense Forces were vaccinated with vaccinia virus (VACV) strain LC16m8, an attenuated smallpox vaccine derived from VACV strain Lister. The serological response induced by LC16m8 to four virion-surface proteins and the intracellular mature virus (IMV) and extracellular enveloped virus (EEV) was investigated. LC16m8 induced antibody response against the IMV protein A27 and the EEV protein A56. LC16m8 also induced IMV-neutralizing antibodies, but unlike the VACV strain Lister, did not induce either EEV-neutralizing antibody or antibody to EEV protein B5, except after revaccination. Given that B5 is the only target for EEV-neutralizing antibody and that neutralization of both IMV and EEV give optimal protection against orthopoxvirus challenge, these data suggest that immunity induced by LC16m8 might be less potent than that deriving from strain Lister. This potential disadvantage should be balanced against the advantage of the greater safety of LC16m8.
Collapse
Affiliation(s)
- Benjamin F Johnson
- Section of Virology, Faculty of Medicine, Imperial College London, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| | - Yasuhiro Kanatani
- National Institute of Public Health, Department of Policy Sciences, 2-3-6 Minami, Wako-shi, Saitama 351-0197, Japan
| | - Tatsuya Fujii
- Self-Defense Force, Central Hospital, Health Department, Management Division, 1-2-24 Ikejiri, Setagaya, Tokyo 154-8532, Japan
| | - Tomoya Saito
- Bio-preparedness Research Laboratory, Department of Tropical Medicine and Parasitology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Hiroyuki Yokote
- The Chemo-Sero-Therapeutic Research Institute (KAKETSUKEN), 1-6-1 Okubo, Kumamoto 860-8568, Japan
| | - Geoffrey L Smith
- Section of Virology, Faculty of Medicine, Imperial College London, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| |
Collapse
|