1
|
Shang C, Shi S, Jiang Q, Wang X, Yao X, Li W, Song G, Li Y, Sun Y, Hu J, Zhang C, Zhu Y, Liu Z, Gu C, Liu Y, Shi W, Zhao Z, Li X. Clinical manifestations and pathogenicity of Clade IIb monkeypox virus in rabbits. Emerg Microbes Infect 2025; 14:2465309. [PMID: 39945750 PMCID: PMC11849023 DOI: 10.1080/22221751.2025.2465309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025]
Abstract
The 2022 monkeypox outbreak involved rapid global dissemination, prompting research into animal models for the monkeypox virus (MPXV), including non-human primates and mice. However, studies utilizing rabbits as models remain limited. In this study, we established three rabbit models using the current epidemic MPXV strain. Following intravenous MPXV injection, adult rabbits exhibited characteristic clinical manifestations, including widespread rash and fever, with viral replication in the skin, lungs, and testes, resulting in severe pathological damage by 6 days post-infection (dpi). Intradermal injection of MPXV into the dorsal skin of adult rabbits produced red lesions with central necrosis and hemorrhage accompanied by dense inflammatory infiltrates. Abundant viral particles were observed in epidermal cells at 6 dpi. Additionally, a fatal MPXV model was developed in 10-day-old rabbits using intranasal virus administration. These young rabbits exhibited lethargy and diarrhea beginning at 2 dpi, significant weight loss, and a 50% mortality rate by 15 dpi. Viral dissemination was detected in multiple organs, leading to extensive multi-organ damage. This study highlights the utility of rabbit models for MPXV, displaying typical clinical features and pathogenic mechanisms.
Collapse
Affiliation(s)
- Chao Shang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Shaowen Shi
- Hebei Agricultural University, Baoding, People’s Republic of China
| | - Qiwei Jiang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Xiaohan Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, People’s Republic of China
| | - Wanzi Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, People’s Republic of China
| | - Gaojie Song
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| | - Yiquan Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Yongyang Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Jinglei Hu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Cuiling Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Yilong Zhu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Zirui Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Chaode Gu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Yan Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Wanyu Shi
- Hebei Agricultural University, Baoding, People’s Republic of China
| | - Zongzheng Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Xiao Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| |
Collapse
|
2
|
Falasca L, Mija C, Sberna G, Francalancia M, Meschi S, Mazzotta V, Girardi E, Antinori A, Maggi F, Bordi L. Antiviral Effects of Tecovirimat and Cellular Ultrastructural Changes in Human Bronchial Epithelial Cell Line Following Monkeypox Virus Infection. Int J Mol Sci 2025; 26:2718. [PMID: 40141361 PMCID: PMC11942983 DOI: 10.3390/ijms26062718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/06/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
The mpox virus (MPXV) Clade IIb outbreak in 2022 was the biggest one ever to occur outside Africa, causing different types of clinical symptoms and levels of disease severity. There is no currently approved treatment for mpox, but Tecovirimat has proven effective against known orthopoxviruses in several animal models and Vero cell cultures. Since serious complications, including lung involvement, have been reported, especially in immunocompromised people, we investigated the effects of MPXV infection on the in vitro model of lung airway epithelium (Calu-3 cell line) and examined MPXV replication kinetic and related ultrastructural changes, also performing dose-response studies to measure Tecovirimat antiviral activity. Our results highlighted an active replication of MPXV in Calu-3 cells linked to mitochondrial structural modifications with perinuclear relocation and the formation of cytoplasmic vacuoles. Treatment with Tecovirimat consistently reduced viral replication both in supernatants (81%) and inside cells (77%) and ultimately stopped viral infectivity (92% of cytopathic effect reduction) after 48 h of infection. Drug administration inhibited the final wrapping of mature viral particles, causing extensive cytoplasmic vacuolation. Our results demonstrated Tecovirimat's in vitro effectiveness against MPXV at the nanomolar concentration on Calu-3 cells. This suggests a potential rationale for using this drug for patients with mpox severe disease and lung involvement.
Collapse
Affiliation(s)
- Laura Falasca
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases Lazzaro Spallanzani—IRCCS, 00149 Rome, Italy;
| | - Cosmina Mija
- Laboratory of Virology and Biosafety Laboratories, National Institute for Infectious Diseases Lazzaro Spallanzani—IRCCS, 00149 Rome, Italy; (C.M.)
| | - Giuseppe Sberna
- Laboratory of Virology and Biosafety Laboratories, National Institute for Infectious Diseases Lazzaro Spallanzani—IRCCS, 00149 Rome, Italy; (C.M.)
| | - Massimo Francalancia
- Microbiology and Virology Unit, San Gallicano Dermatological Institute—IRCCS, Istituti Fisioterapici Ospitalieri (IFO), 00144 Rome, Italy
| | - Silvia Meschi
- Laboratory of Virology and Biosafety Laboratories, National Institute for Infectious Diseases Lazzaro Spallanzani—IRCCS, 00149 Rome, Italy; (C.M.)
| | - Valentina Mazzotta
- Clinical Department, National Institute for Infectious Diseases Lazzaro Spallanzani—IRCCS, 00149 Rome, Italy; (V.M.); (A.A.)
| | - Enrico Girardi
- Scientific Direction, National Institute for Infectious Diseases Lazzaro Spallanzani—IRCCS, 00149 Rome, Italy
| | - Andrea Antinori
- Clinical Department, National Institute for Infectious Diseases Lazzaro Spallanzani—IRCCS, 00149 Rome, Italy; (V.M.); (A.A.)
| | - Fabrizio Maggi
- Laboratory of Virology and Biosafety Laboratories, National Institute for Infectious Diseases Lazzaro Spallanzani—IRCCS, 00149 Rome, Italy; (C.M.)
| | - Licia Bordi
- Laboratory of Virology and Biosafety Laboratories, National Institute for Infectious Diseases Lazzaro Spallanzani—IRCCS, 00149 Rome, Italy; (C.M.)
| |
Collapse
|
3
|
Luo YH, Zhang T, Cao JL, Hou WS, Wang AQ, Jin CH. Monkeypox: An outbreak of a rare viral disease. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024; 57:1-10. [PMID: 38177001 DOI: 10.1016/j.jmii.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/24/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Monkeypox is a viral zoonotic disease rarely found outside Africa. Monkeypox can be spread from person to person through close contact with an infected person, and the rate of transmission is not very high. In addition, monkeypox and variola virus are both pox viruses, and the spread of monkeypox virus was also controlled to some extent by the smallpox campaign, so monkeypox was not widely paid attention to. However, as smallpox vaccination is phased out in various countries or regions, people's resistance to orthopoxviruses is decreasing, especially among people who have not been vaccinated against smallpox. This has led to a significant increase in the frequency and geographical distribution of human monkeypox cases in recent years, and the monkeypox virus has become the orthopoxvirus that poses the greatest threat to public health. Since the last large-scale monkeypox infection was detected in 2022, the number of countries or territories affected has exceeded 100. Many confirmed and suspected cases of monkeypox have been found in individuals who have not travelled to affected areas, and the route of infection is not obvious, making this outbreak of monkeypox a cause for concern globally. The purpose of this systematic review is to further understand the pathophysiological and epidemiological characteristics of monkeypox, as well as existing prevention and treatment methods, with a view to providing evidence for the control of monkeypox.
Collapse
Affiliation(s)
- Ying-Hua Luo
- Department of Grass Science, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Tong Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Jing-Long Cao
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Wen-Shuang Hou
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - An-Qi Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China; National Coarse Cereals Engineering Research Center, Daqing, 163319, China.
| |
Collapse
|
4
|
Smith TG, Gigante CM, Wynn NT, Matheny A, Davidson W, Yang Y, Condori RE, O'Connell K, Kovar L, Williams TL, Yu YC, Petersen BW, Baird N, Lowe D, Li Y, Satheshkumar PS, Hutson CL. Tecovirimat Resistance in Mpox Patients, United States, 2022-2023. Emerg Infect Dis 2023; 29:2426-2432. [PMID: 37856204 PMCID: PMC10683829 DOI: 10.3201/eid2912.231146] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023] Open
Abstract
During the 2022 multinational outbreak of monkeypox virus (MPXV) infection, the antiviral drug tecovirimat (TPOXX; SIGA Technologies, Inc., https://www.siga.com) was deployed in the United States on a large scale for the first time. The MPXV F13L gene homologue encodes the target of tecovirimat, and single amino acid changes in F13 are known to cause resistance to tecovirimat. Genomic sequencing identified 11 mutations previously reported to cause resistance, along with 13 novel mutations. Resistant phenotype was determined using a viral cytopathic effect assay. We tested 124 isolates from 68 patients; 96 isolates from 46 patients were found to have a resistant phenotype. Most resistant isolates were associated with severely immunocompromised mpox patients on multiple courses of tecovirimat treatment, whereas most isolates identified by routine surveillance of patients not treated with tecovirimat remained sensitive. The frequency of resistant viruses remains relatively low (<1%) compared with the total number of patients treated with tecovirimat.
Collapse
|
5
|
Russo AT, Grosenbach DW, Honeychurch KM, Long PG, Hruby DE. Overview of the regulatory approval of tecovirimat intravenous formulation for treatment of smallpox: potential impact on smallpox outbreak response capabilities, and future tecovirimat development potential. Expert Rev Anti Infect Ther 2023; 21:235-242. [PMID: 36728515 PMCID: PMC10054055 DOI: 10.1080/14787210.2023.2170350] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/16/2023] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Tecovirimat oral capsule formulation is approved in the US and Canada for treatment of smallpox and in the United Kingdom (UK) and European Union (EU) for treatment of multiple human orthopoxvirus diseases, including mpox. Smallpox is considered a serious threat, and there is currently an unprecedented global mpox outbreak. AREAS COVERED A brief summary of the threat of smallpox, the threat of increasing mpox spread in endemic regions, and the unprecedented emergence of mpox into non-endemic regions is presented. The tecovirimat intravenous formulation clinical development program leading to USFDA approval for smallpox treatment is discussed. EXPERT OPINION As of January 2023 tecovirimat is approved to treat mpox in the UK and EU. However, published clinical trial data evaluating tecovirimat efficacy and safety in mpox patients is pending. Increasing global prevalence of mpox highlights the potential benefits of a well-characterized, effective, and safe antiviral treatment for mpox infection. Ongoing trials in mpox patients may provide results supporting the use of tecovirimat to treat this disease. USFDA approval of tecovirimat for post-exposure prophylaxis in the event of a smallpox release, and the development of pediatric liquid formulations for patients under 13 kg, could provide additional public health benefits.
Collapse
|
6
|
Mpox: Special Considerations in the Immunocompromised Host. CURRENT TREATMENT OPTIONS IN INFECTIOUS DISEASES 2023. [DOI: 10.1007/s40506-022-00260-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
7
|
Byareddy SN, Sharma K, Sachdev S, Reddy AS, Acharya A, Klaustermeier KM, Lorson CL, Singh K. Potential therapeutic targets for Mpox: the evidence to date. Expert Opin Ther Targets 2023; 27:419-431. [PMID: 37368464 PMCID: PMC10722886 DOI: 10.1080/14728222.2023.2230361] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/07/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
INTRODUCTION The global Mpox (MPX) disease outbreak caused by the Mpox virus (MPXV) in 2022 alarmed the World Health Organization (WHO) and health regulation agencies of individual countries leading to the declaration of MPX as a Public Health Emergency. Owing to the genetic similarities between smallpox-causing poxvirus and MPXV, vaccine JYNNEOS, and anti-smallpox drugs Brincidofovir and Tecovirimat were granted emergency use authorization by the United States Food and Drug Administration. The WHO also included cidofovir, NIOCH-14, and other vaccines as treatment options. AREAS COVERED This article covers the historical development of EUA-granted antivirals, resistance to these antivirals, and the projected impact of signature mutations on the potency of antivirals against currently circulating MPXV. Since a high prevalence of MPXV infections in individuals coinfected with HIV and MPXV, the treatment results among these individuals have been included. EXPERT OPINION All EUA-granted drugs have been approved for smallpox treatment. These antivirals show good potency against Mpox. However, conserved resistance mutation positions in MPXV and related poxviruses, and the signature mutations in the 2022 MPXV can potentially compromise the efficacy of the EUA-granted treatments. Therefore, MPXV-specific medications are required not only for the current but also for possible future outbreaks.
Collapse
Affiliation(s)
- Siddappa N Byareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | - Shrikesh Sachdev
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Athreya S. Reddy
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | - Christian L Lorson
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Kamal Singh
- Department of Pharmaceutical Chemistry, DPSRU, New Delhi-110017
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
8
|
Abstract
Human monkeypox is a viral zoonosis endemic to West and Central Africa that has recently generated increased interest and concern on a global scale as an emerging infectious disease threat in the midst of the slowly relenting COVID-2019 disease pandemic. The hallmark of infection is the development of a flu-like prodrome followed by the appearance of a smallpox-like exanthem. Precipitous person-to-person transmission of the virus among residents of 100 countries where it is nonendemic has motivated the immediate and widespread implementation of public health countermeasures. In this review, we discuss the origins and virology of monkeypox virus, its link with smallpox eradication, its record of causing outbreaks of human disease in regions where it is endemic in wildlife, its association with outbreaks in areas where it is nonendemic, the clinical manifestations of disease, laboratory diagnostic methods, case management, public health interventions, and future directions.
Collapse
Affiliation(s)
- Sameer Elsayed
- Department of Medicine, Western University, London, Ontario, Canada
- Department of Pathology & Laboratory Medicine, Western University, London, Ontario, Canada
- Department of Epidemiology & Biostatistics, Western University, London, Ontario, Canada
| | - Lise Bondy
- Department of Medicine, Western University, London, Ontario, Canada
| | - William P. Hanage
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Sukhdeo S, Mishra S, Walmsley S. Human monkeypox: a comparison of the characteristics of the new epidemic to the endemic disease. BMC Infect Dis 2022; 22:928. [PMID: 36503476 PMCID: PMC9742013 DOI: 10.1186/s12879-022-07900-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022] Open
Abstract
In May 2022, a new global outbreak of mpox (formerly, human monkeypox) emerged that was declared a public health emergency of international concern by the World Health Organization on July 23, 2022. With new patterns of person-to-person spread within sexual networks in nonendemic countries and several differences from the classic disease course, we performed a comprehensive review of existing literature on human monkeypox to discuss epidemiology, modes of transmission, clinical presentation and asymptomatic infection, diagnostics, therapeutics, and vaccines with the primary aim to identify important areas for future research of this new epidemic form of the disease. A comprehensive literature search was performed of all published literature to August 15, 2022. Historically, in regions of monkeypox virus endemicity, human outbreaks have occurred related to discrete zoonotic events. The animal reservoir is unknown, but the virus has been isolated from rodents. Traditionally, transmission occurred by direct or indirect contact with an infected animal. In nonendemic countries affected in the 2022 outbreak, almost exclusive person-to-person spread has been observed, and most cases are connected to sexual networks of gay, bisexual, and other men who have sex with men. After an incubation period of approximately 13 days, in traditional human cases affected persons developed a febrile prodrome preceding a rash that started on the face and body, spread centrifugally to the palms and soles and healed monomorphically over two to four weeks. However, in the 2022 outbreak, the febrile illness is often absent or occurs after the onset of the rash. The rash presents primarily in the anogenital region and face before disseminating throughout the body, with lesions displaying regional pleomorphism. There is a paucity of data for the role of antiviral agents or vaccines. The epidemiology and clinical course of mpox has changed in the 2022 epidemic from that observed with the endemic disease. There is an urgent need to establish rapid and collaborative research platforms to diagnose, treat and prevent disease and inform important public health and other strategies to stop the spread of disease.
Collapse
Affiliation(s)
- Sharon Sukhdeo
- Department of Medicine, University of Toronto, Toronto, Canada.
| | - Sharmistha Mishra
- Division of Infectious Diseases, Department of Medicine, St. Michael's Hospital, MAP Centre for Urban Health Solutions, University of Toronto, Toronto, Canada
| | - Sharon Walmsley
- Department of Medicine, Division of Infectious Diseases, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
10
|
Parker S, D'Angelo J, Buller RM, Smee DF, Lantto J, Nielsen H, Jensen A, Prichard M, George SL. A human recombinant analogue to plasma-derived vaccinia immunoglobulin prophylactically and therapeutically protects against lethal orthopoxvirus challenge. Antiviral Res 2021; 195:105179. [PMID: 34530009 PMCID: PMC9628779 DOI: 10.1016/j.antiviral.2021.105179] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 11/22/2022]
Abstract
Orthopoxviruses such as variola and monkeypox viruses continue to threaten the human population. Monkeypox virus is endemic in central and western Africa and outbreaks have reached as far as the U.S. Although variola virus, the etiologic agent of smallpox, has been eradicated by a successful vaccination program, official and likely clandestine stocks of the virus exist. Moreover, studies with ectromelia virus (the etiological agent of mousepox) have revealed that IL-4 recombinant viruses are significantly more virulent than wild-type viruses even in mice treated with vaccines and/or antivirals. For these reasons, it is critical that antiviral modalities are developed to treat these viruses should outbreaks, or deliberate dissemination, occur. Currently, 2 antivirals (brincidofovir and tecovirimat) are in the U.S. stockpile allowing for emergency use of the drugs to treat smallpox. Both antivirals have advantages and disadvantages in a clinical and emergency setting. Here we report on the efficacy of a recombinant immunoglobulin (rVIG) that demonstrated efficacy against several orthopoxviruses in vitro and in vivo in both a prophylactic and therapeutic fashion. A single intraperitoneal injection of rVIG significantly protected mice when given up to 14 days before or as late as 6 days post challenge. Moreover, rVIG reduced morbidity, as measured by weight-change, as well as several previously established biomarkers of disease. In rVIG treated mice, we found that vDNA levels in blood were significantly reduced, as was ALT (a marker of liver damage) and infectious virus levels in the liver. No apparent adverse events were observed in rVIG treated mice, suggesting the immunoglobulin is well tolerated. These findings suggest that recombinant immunoglobulins could be candidates for further evaluation and possible licensure under the FDA Animal Rule.
Collapse
Affiliation(s)
- Scott Parker
- Division of Infectious Diseases, Department of Internal Medicine, Saint Louis University, and St. Louis VA Medical Center, St. Louis, MO, 63104, USA
| | - June D'Angelo
- Division of Infectious Diseases, Department of Internal Medicine, Saint Louis University, and St. Louis VA Medical Center, St. Louis, MO, 63104, USA
| | - R Mark Buller
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, 63104, USA
| | - Donald F Smee
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, 84322, USA
| | - Johan Lantto
- Symphogen, Pederstrupvej 93, DK-2750, Ballerup, Denmark
| | | | - Allan Jensen
- Symphogen, Pederstrupvej 93, DK-2750, Ballerup, Denmark
| | - Mark Prichard
- Department of Pediatrics, University of Alabama, Birmingham, AL, 35233, USA
| | - Sarah L George
- Division of Infectious Diseases, Department of Internal Medicine, Saint Louis University, and St. Louis VA Medical Center, St. Louis, MO, 63104, USA.
| |
Collapse
|
11
|
Suresh MK, Vasudevan AK, Biswas L, Biswas R. Protective efficacy of Alum adjuvanted Amidase protein vaccine against Staphylococcus aureus infection in multiple mouse models. J Appl Microbiol 2021; 132:1422-1434. [PMID: 34487603 DOI: 10.1111/jam.15291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 08/05/2021] [Accepted: 08/23/2021] [Indexed: 12/20/2022]
Abstract
AIMS Staphylococcus aureus is an opportunistic pathogen of humans. No commercial vaccine is available to combat S. aureus infections. In this study, we have investigated the protective immune response generated by S. aureus non-covalently associated cell wall surface protein N-acetylmuramoyl-L-alanine amidase (AM) in combination with Alum (Al) and heat-killed S. aureus (hkSA) using murine models. METHODS AND RESULTS BALB/c mice were immunized with increasing concentrations of AM antigen or hkSA to determine their optimum concentration for vaccination. Fifty micrograms of AM and hkSA each were found to generate maximum anti-AM IgG antibody production. BALB/c mice were immunized next with 50 µg of AM, 50 µg of hKSA and 1 mg Al vaccine formulation. Vaccine efficacy was validated by challenging immunized BALB/c mice with S. aureus Newman and three clinical methicillin-resistant S. aureus strains. AM-hkSA-Al-immunized mice generated high anti-AM IgG antibody response with IgG1 and IgG2b as the predominant immunoglobulin subtypes. Increased survival (60%-90%) with decreased clinical disease symptoms was observed in the vaccinated BALB/c mice group. A significantly lower bacterial load and decreased kidney abscess formation was observed following the challenge with S. aureus in the vaccinated BALB/c mice group. Furthermore, the efficacy of AM-hkSA-Al vaccine was also validated using C57 BL/6 and Swiss albino mice. CONCLUSIONS Using murine infection models, we have demonstrated that AM-hkSA-Al vaccine would be effective in preventing S. aureus infections. SIGNIFICANCE AND IMPACT OF STUDY AM-hkSA-Al vaccine elicited strong immune response and may be considered for future vaccine design against S. aureus infections.
Collapse
Affiliation(s)
- Maneesha K Suresh
- Center for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Anil Kumar Vasudevan
- Department of Microbiology, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, AIMS - Ponekkara, Cochin, India
| | - Lalitha Biswas
- Center for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Raja Biswas
- Center for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| |
Collapse
|
12
|
Russo AT, Grosenbach DW, Chinsangaram J, Honeychurch KM, Long PG, Lovejoy C, Maiti B, Meara I, Hruby DE. An overview of tecovirimat for smallpox treatment and expanded anti-orthopoxvirus applications. Expert Rev Anti Infect Ther 2020; 19:331-344. [PMID: 32882158 PMCID: PMC9491074 DOI: 10.1080/14787210.2020.1819791] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction Tecovirimat (TPOXX®; ST-246) was approved for the treatment of symptomatic smallpox by the USFDA in July of 2018 and has been stockpiled by the US government for use in a smallpox outbreak. While there has not been a reported case of smallpox since 1978 it is still considered a serious bioterrorism threat. Areas covered A brief history of smallpox from its proposed origins as a human disease through its eradication in the late 20th century is presented. The current smallpox threat and the current public health response plans are described. The discovery, and development of tecovirimat through NDA submission and subsequent approval for treatment of smallpox are discussed. Google Scholar and PubMed were searched over all available dates for relevant publications. Expert opinion Approval of tecovirimat to treat smallpox represents an important milestone in biosecurity preparedness. Incorporating tecovirimat into the CDC smallpox response plan, development of pediatric liquid and intravenous formulations, and approval for post-exposure prophylaxis would provide additional health security benefit. Tecovirimat shows broad efficacy against orthopoxviruses in vitro and in vivo and could be developed for use against emerging orthopoxvirus diseases such as monkeypox, vaccination-associated adverse events, and side effects of vaccinia oncolytic virus therapy.
Collapse
Affiliation(s)
- Andrew T Russo
- Poxvirus Research Group, SIGA Technologies, Inc, Corvallis, OR, USA
| | | | | | | | - Paul G Long
- Regulatory Affairs, SIGA Technologies, Inc, Corvallis, OR, USA
| | - Candace Lovejoy
- Program Management, SIGA Technologies, Inc, Corvallis, OR, USA
| | - Biswajit Maiti
- Drug Metabolism & Pharmacokinetics, SIGA Technologies, Inc, Corvallis, OR, USA
| | - Ingrid Meara
- Clinical Research, SIGA Technologies, Inc, Corvallis, OR, USA
| | - Dennis E Hruby
- Chief Scientific Officer, SIGA Technologies, Inc, Corvallis, OR, USA
| |
Collapse
|
13
|
Priyamvada L, Alabi P, Leon A, Kumar A, Sambhara S, Olson VA, Sello JK, Satheshkumar PS. Discovery of Retro-1 Analogs Exhibiting Enhanced Anti-vaccinia Virus Activity. Front Microbiol 2020; 11:603. [PMID: 32390964 PMCID: PMC7190985 DOI: 10.3389/fmicb.2020.00603] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/18/2020] [Indexed: 11/13/2022] Open
Abstract
Orthopoxviruses (OPXVs) are an increasing threat to human health due to the growing population of OPXV-naive individuals after the discontinuation of routine smallpox vaccination. Antiviral drugs that are effective as postexposure treatments against variola virus (the causative agent of smallpox) or other OPXVs are critical in the event of an OPXV outbreak or exposure. The only US Food and Drug Administration-approved drug to treat smallpox, Tecovirimat (ST-246), exerts its antiviral effect by inhibiting extracellular virus (EV) formation, thereby preventing cell-cell and long-distance spread. We and others have previously demonstrated that host Golgi-associated retrograde proteins play an important role in monkeypox virus (MPXV) and vaccinia virus (VACV) EV formation. Inhibition of the retrograde pathway by small molecules such as Retro-2 has been shown to decrease VACV infection in vitro and to a lesser extent in vivo. To identify more potent inhibitors of the retrograde pathway, we screened a large panel of compounds containing a benzodiazepine scaffold like that of Retro-1, against VACV infection. We found that a subset of these compounds displayed better anti-VACV activity, causing a reduction in EV particle formation and viral spread compared to Retro-1. PA104 emerged as the most potent analog, inhibiting 90% viral spread at 1.3 μM with a high selectivity index. In addition, PA104 strongly inhibited two distinct ST-246-resistant viruses, demonstrating its potential benefit for use in combination therapy with ST-246. These data and further characterizations of the specific protein targets and in vivo efficacy of PA104 may have important implications for the design of effective antivirals against OPXV.
Collapse
Affiliation(s)
- Lalita Priyamvada
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Philip Alabi
- Department of Chemistry, Brown University, Providence, RI, United States
| | - Andres Leon
- Department of Chemistry, Brown University, Providence, RI, United States
| | - Amrita Kumar
- Immunology and Pathogenesis Branch, Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Suryaprakash Sambhara
- Immunology and Pathogenesis Branch, Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Victoria A Olson
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jason K Sello
- Department of Chemistry, Brown University, Providence, RI, United States
| | | |
Collapse
|
14
|
Merchlinsky M, Albright A, Olson V, Schiltz H, Merkeley T, Hughes C, Petersen B, Challberg M. The development and approval of tecoviromat (TPOXX ®), the first antiviral against smallpox. Antiviral Res 2019; 168:168-174. [PMID: 31181284 DOI: 10.1016/j.antiviral.2019.06.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 06/06/2019] [Indexed: 11/16/2022]
Abstract
The classification of smallpox by the U.S. Centers for Disease Control and Prevention (CDC) as a Category A Bioterrorism threat agent has resulted in the U.S. Government investing significant funds to develop and stockpile a suite of medical countermeasures to ameliorate the consequences of a smallpox epidemic. This stockpile includes both vaccines for prophylaxis and antivirals to treat symptomatic patients. In this manuscript, we describe the path to approval for the first therapeutic against smallpox, identified during its development as ST-246, now known as tecovirimat and TPOXX®, a small-molecule antiviral compound sponsored by SIGA Technologies to treat symptomatic smallpox. Because the disease is no longer endemic, the development and approval of TPOXX® was only possible under the U.S. Food and Drug and Administration Animal Rule (FDA 2002). In this article, we describe the combination of animal model studies and clinical trials that were used to satisfy the FDA requirements for the approval of TPOXX ® under the Animal Rule.
Collapse
Affiliation(s)
- Michael Merchlinsky
- Biomedical Advanced Research and Development Authority, 300 C Street SW, Washington DC, 20201, USA.
| | - Andrew Albright
- Biomedical Advanced Research and Development Authority, 300 C Street SW, Washington DC, 20201, USA
| | - Victoria Olson
- National Center for Emerging and Zoonotic Infectious Disease, Centers for Disease Control and Prevention, Mail Stop G-06, 1600 Clifton Road, NE, Atlanta, 30333, Georgia
| | - Helen Schiltz
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, MSC 9825, 5601 Fishers Lane, Rockville, MD, 20851, USA
| | - Tyler Merkeley
- Biomedical Advanced Research and Development Authority, 300 C Street SW, Washington DC, 20201, USA
| | - Claiborne Hughes
- Biomedical Advanced Research and Development Authority, 300 C Street SW, Washington DC, 20201, USA
| | - Brett Petersen
- National Center for Emerging and Zoonotic Infectious Disease, Centers for Disease Control and Prevention, Mail Stop G-06, 1600 Clifton Road, NE, Atlanta, 30333, Georgia
| | - Mark Challberg
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, MSC 9825, 5601 Fishers Lane, Rockville, MD, 20851, USA
| |
Collapse
|
15
|
Russo AT, Grosenbach DW, Brasel TL, Baker RO, Cawthon AG, Reynolds E, Bailey T, Kuehl PJ, Sugita V, Agans K, Hruby DE. Effects of Treatment Delay on Efficacy of Tecovirimat Following Lethal Aerosol Monkeypox Virus Challenge in Cynomolgus Macaques. J Infect Dis 2018; 218:1490-1499. [PMID: 29982575 PMCID: PMC6151088 DOI: 10.1093/infdis/jiy326] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/15/2018] [Indexed: 12/17/2022] Open
Abstract
Background Tecovirimat (ST-246) is being developed as an antiviral therapeutic for smallpox for use in the event of an accidental or intentional release. The last reported case of smallpox was 1978 but the potential for use of variola virus for biowarfare has renewed interest in smallpox antiviral therapeutics. Methods Cynomolgus macaques were challenged with a lethal dose of monkeypox virus (MPXV) by aerosol as a model for human smallpox and treated orally with 10 mg/kg tecovirimat once daily starting up to 8 days following challenge. Monkeys were monitored for survival, lesions, and clinical signs of disease. Samples were collected for measurement of viremia by quantitative real-time polymerase chain reaction, and for white blood cell counts. Results Survival in animals initiating treatment up to 5 days postchallenge was 100%. In animals treated starting 6, 7, or 8 days following challenge, survival was 67%, 100%, and 50%, respectively. Treatment initiation up to 4 days following challenge reduced severity of clinical manifestations of infection. Conclusions Tecovirimat treatment initiated up to 8 days following a lethal aerosol MPXV challenge improves survival and, when initiated earlier than 5 days after challenge, provides protection from clinical effects of disease, supporting the conclusion that it is a promising smallpox antiviral therapeutic candidate.
Collapse
Affiliation(s)
- Andrew T Russo
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- Poxvirus Research Group, SIGA Technologies, Inc, Corvallis, Oregon
| | | | - Trevor L Brasel
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston
| | - Robert O Baker
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- Microbiology and Molecular Biology Division, Illinois Institute of Technology Research Institute, Chicago
| | - Andrew G Cawthon
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- Bacteriology, Virology and In Vitro Operations, Battelle Memorial Institute, Columbus, Ohio
| | - Erin Reynolds
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- Department of Pathology, University of Texas Medical Branch, Galveston
| | - Tara Bailey
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- Covance Laboratories, Madison, Wisconsin
| | - Philip J Kuehl
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Victoria Sugita
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- University of New Mexico, Albuquerque
| | - Krystle Agans
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston
| | | |
Collapse
|
16
|
Pires MA, Rodrigues NFS, de Oliveira DB, de Assis FL, Costa GB, Kroon EG, Mota BEF. In vitro susceptibility to ST-246 and Cidofovir corroborates the phylogenetic separation of Brazilian Vaccinia virus into two clades. Antiviral Res 2018; 152:36-44. [PMID: 29427676 DOI: 10.1016/j.antiviral.2018.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/29/2018] [Accepted: 02/05/2018] [Indexed: 01/01/2023]
Abstract
The Orthopoxvirus (OPV) genus of the Poxviridae family contains several human pathogens, including Vaccinia virus (VACV), which have been implicating in outbreaks of a zoonotic disease called Bovine Vaccinia in Brazil. So far, no approved treatment exists for OPV infections, but ST-246 and Cidofovir (CDV) are now in clinical development. Therefore, the objective of this work was to evaluate the susceptibility of five strains of Brazilian VACV (Br-VACV) to ST-246 and Cidofovir. The susceptibility of these strains to both drugs was evaluated by plaque reduction assay, extracellular virus's quantification in the presence of ST-246 and one-step growth curve in cells treated with CDV. Besides that, the ORFs F13L and E9L were sequenced for searching of polymorphisms associated with drug resistance. The effective concentration of 50% (EC50) from both drugs varies significantly for different strains (from 0.0054 to 0.051 μM for ST-246 and from 27.14 to 61.23 μM for CDV). ST-246 strongly inhibits the production of extracellular virus for all isolates in concentrations as low as 0.1 μM and it was observed a relevant decrease of progeny production for all Br-VACV after CDV treatment. Sequencing of the F13L and E9L ORFs showed that Br-VACV do not present the polymorphism(s) associated with resistance to ST-246 and CDV. Taken together, our results showed that ST-246 and CDV are effective against diverse, wild VACV strains and that the susceptibility of Br-VACV to these drugs mirrored the phylogenetic split of these isolates into two groups. Thus, both ST-246 and CDV are of great interest as compounds to treat individuals during Bovine Vaccinia outbreaks in Brazil.
Collapse
Affiliation(s)
- Mariana A Pires
- Laboratório de Microbiologia Clínica, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP 31270-901, Belo Horizonte, Brazil
| | - Nathália F S Rodrigues
- Laboratório de Microbiologia Clínica, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP 31270-901, Belo Horizonte, Brazil
| | - Danilo B de Oliveira
- Laboratório de Vírus, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP 31270-901, Belo Horizonte, Brazil
| | - Felipe L de Assis
- Laboratório de Vírus, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP 31270-901, Belo Horizonte, Brazil
| | - Galileu B Costa
- Laboratório de Vírus, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP 31270-901, Belo Horizonte, Brazil
| | - Erna G Kroon
- Laboratório de Vírus, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP 31270-901, Belo Horizonte, Brazil
| | - Bruno E F Mota
- Laboratório de Microbiologia Clínica, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, CEP 31270-901, Belo Horizonte, Brazil.
| |
Collapse
|
17
|
Trindade GDS, Emerson GL, Sammons S, Frace M, Govil D, Fernandes Mota BE, Abrahão JS, de Assis FL, Olsen-Rasmussen M, Goldsmith CS, Li Y, Carroll D, Guimarães da Fonseca F, Kroon E, Damon IK. Serro 2 Virus Highlights the Fundamental Genomic and Biological Features of a Natural Vaccinia Virus Infecting Humans. Viruses 2016; 8:v8120328. [PMID: 27973399 PMCID: PMC5192389 DOI: 10.3390/v8120328] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/01/2016] [Accepted: 11/24/2016] [Indexed: 01/14/2023] Open
Abstract
Vaccinia virus (VACV) has been implicated in infections of dairy cattle and humans, and outbreaks have substantially impacted local economies and public health in Brazil. During a 2005 outbreak, a VACV strain designated Serro 2 virus (S2V) was collected from a 30-year old male milker. Our aim was to phenotypically and genetically characterize this VACV Brazilian isolate. S2V produced small round plaques without associated comets when grown in BSC40 cells. Furthermore, S2V was less virulent than the prototype strain VACV-Western Reserve (WR) in a murine model of intradermal infection, producing a tiny lesion with virtually no surrounding inflammation. The genome of S2V was sequenced by primer walking. The coding region spans 184,572 bp and contains 211 predicted genes. Mutations in envelope genes specifically associated with small plaque phenotypes were not found in S2V; however, other alterations in amino acid sequences within these genes were identified. In addition, some immunomodulatory genes were truncated in S2V. Phylogenetic analysis using immune regulatory-related genes, besides the hemagglutinin gene, segregated the Brazilian viruses into two clusters, grouping the S2V into Brazilian VACV group 1. S2V is the first naturally-circulating human-associated VACV, with a low passage history, to be extensively genetically and phenotypically characterized.
Collapse
Affiliation(s)
- Giliane de Souza Trindade
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, MG CEP 31270-901, Brazil.
| | - Ginny L Emerson
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Scott Sammons
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Michael Frace
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Dhwani Govil
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | | | - Jônatas Santos Abrahão
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, MG CEP 31270-901, Brazil.
| | - Felipe Lopes de Assis
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, MG CEP 31270-901, Brazil.
| | - Melissa Olsen-Rasmussen
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Cynthia S Goldsmith
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Yu Li
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | - Darin Carroll
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| | | | - Erna Kroon
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, MG CEP 31270-901, Brazil.
| | - Inger K Damon
- Coordinating Center for Infectious Diseases, Centers for Disease Control and Prevention (CCID/CDC), Atlanta, 30329-4027 GA, USA.
| |
Collapse
|
18
|
Klimochkin YN, Shiryaev VA, Leonova MV. Antiviral properties of cage compounds. New prospects. Russ Chem Bull 2016. [DOI: 10.1007/s11172-015-1035-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Liu Q, Fan C, Zhou S, Guo Y, Zuo Q, Ma J, Liu S, Wu X, Peng Z, Fan T, Guo C, Shen Y, Huang W, Li B, He Z, Wang Y. Bioluminescent imaging of vaccinia virus infection in immunocompetent and immunodeficient rats as a model for human smallpox. Sci Rep 2015; 5:11397. [PMID: 26235050 PMCID: PMC4522659 DOI: 10.1038/srep11397] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/11/2015] [Indexed: 01/23/2023] Open
Abstract
Due to the increasing concern of using smallpox virus as biological weapons for terrorist attack, there is renewed interest in studying the pathogenesis of human smallpox and development of new therapies. Animal models are highly demanded for efficacy and safety examination of new vaccines and therapeutic drugs. Here, we demonstrated that both wild type and immunodeficient rats infected with an engineered vaccinia virus carrying Firefly luciferase reporter gene (rTV-Fluc) could recapitulate infectious and clinical features of human smallpox. Vaccinia viral infection in wild type Sprague-Dawley (SD) rats displayed a diffusible pattern in various organs, including liver, head and limbs. The intensity of bioluminescence generated from rTV-Fluc correlated well with viral loads in tissues. Moreover, neutralizing antibodies had a protective effect against virus reinfection. The recombination activating gene 2 (Rag2) knockout rats generated by transcription activator-like effector nucleases (TALENs) technology were further used to examine the infectivity of the rTV-Fluc in immunodeficient populations. Here we demonstrated that Rag2-/- rats were more susceptible to rTV-Fluc than SD rats with a slower virus clearance rate. Therefore, the rTV-Fluc/SD rats and rTV-Fluc/Rag2-/- rats are suitable visualization models, which recapitulate wild type or immunodeficient populations respectively, for testing human smallpox vaccine and antiviral drugs.
Collapse
Affiliation(s)
- Qiang Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Changfa Fan
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Shuya Zhou
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Yanan Guo
- Biocytogen Co., Ltd, Beijing, 101111, China
| | - Qin Zuo
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Jian Ma
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Susu Liu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Xi Wu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Zexu Peng
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Tao Fan
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | | | | | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Baowen Li
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Zhengming He
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing, 100050, China
| |
Collapse
|
20
|
Postchallenge administration of brincidofovir protects healthy and immune-deficient mice reconstituted with limited numbers of T cells from lethal challenge with IHD-J-Luc vaccinia virus. J Virol 2015; 89:3295-307. [PMID: 25589648 DOI: 10.1128/jvi.03340-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Protection from lethality by postchallenge administration of brincidofovir (BCV, CMX001) was studied in normal and immune-deficient (nude, nu/nu) BALB/c mice infected with vaccinia virus (VACV). Whole-body bioluminescence imaging was used to record total fluxes in the nasal cavity, lungs, spleen, and liver and to enumerate pox lesions on tails of mice infected via the intranasal route with 10(5) PFU of recombinant IHD-J-Luc VACV expressing luciferase. Areas under the flux curve (AUCs) were calculated for individual mice to assess viral loads. A three-dose regimen of 20 mg/kg BCV administered every 48 h starting either on day 1 or day 2 postchallenge protected 100% of mice. Initiating BCV treatment earlier was more efficient in reducing viral loads and in providing protection from pox lesion development. All BCV-treated mice that survived challenge were also protected from rechallenge with IHD-J-Luc or WRvFire VACV without additional treatment. In immune-deficient mice, BCV protected animals from lethality and reduced viral loads while animals were on the drug. Viral recrudescence occurred within 4 to 9 days, and mice succumbed ∼10 to 20 days after treatment termination. Nude mice reconstituted with 10(5) T cells prior to challenge with 10(4) PFU of IHD-J-Luc and treated with BCV postchallenge survived the infection, cleared the virus from all organs, and survived rechallenge with 10(5) PFU of IHD-J-Luc VACV without additional BCV treatment. Together, these data suggest that BCV protects immunocompetent and partially T cell-reconstituted immune-deficient mice from lethality, reduces viral dissemination in organs, prevents pox lesion development, and permits generation of VACV-specific memory. IMPORTANCE Mass vaccination is the primary element of the public health response to a smallpox outbreak. In addition to vaccination, however, antiviral drugs are required for individuals with uncertain exposure status to smallpox or for whom vaccination is contraindicated. Whole-body bioluminescence imaging was used to study the effect of brincidofovir (BCV) in normal and immune-deficient (nu/nu) mice infected with vaccinia virus, a model of smallpox. Postchallenge administration of 20 mg/kg BCV rescued normal and immune-deficient mice partially reconstituted with T cells from lethality and significantly reduced viral loads in organs. All BCV-treated mice that survived infection were protected from rechallenge without additional treatment. In immune-deficient mice, BCV extended survival. The data show that BCV controls viral replication at the site of challenge and reduces viral dissemination to internal organs, thus providing a shield for the developing adaptive immunity that clears the host of virus and builds virus-specific immunological memory.
Collapse
|
21
|
Graeden E, Fielding R, Steinhouse KE, Rubin IN. Modeling the Effect of Herd Immunity and Contagiousness in Mitigating a Smallpox Outbreak. Med Decis Making 2014; 35:648-59. [DOI: 10.1177/0272989x14561681] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 10/18/2014] [Indexed: 01/13/2023]
Abstract
The smallpox antiviral tecovirimat has recently been purchased by the U.S. Strategic National Stockpile. Given significant uncertainty regarding both the contagiousness of smallpox in a contemporary outbreak and the efficiency of a mass vaccination campaign, vaccine prophylaxis alone may be unable to control a smallpox outbreak following a bioterror attack. Here, we present the results of a compartmental epidemiological model that identifies conditions under which tecovirimat is required to curtail the epidemic by exploring how the interaction between contagiousness and prophylaxis coverage of the affected population affects the ability of the public health response to control a large-scale smallpox outbreak. Each parameter value in the model is based on published empirical data. We describe contagiousness parametrically using a novel method of distributing an assumed R-value over the disease course based on the relative rates of daily viral shedding from human and animal studies of cognate orthopoxvirus infections. Our results suggest that vaccination prophylaxis is sufficient to control the outbreak when caused either by a minimally contagious virus or when a very high percentage of the population receives prophylaxis. As vaccination coverage of the affected population decreases below 70%, vaccine prophylaxis alone is progressively less capable of controlling outbreaks, even those caused by a less contagious virus (R0 less than 4). In these scenarios, tecovirimat treatment is required to control the outbreak (total number of cases under an order of magnitude more than the number of initial infections). The first study to determine the relative importance of smallpox prophylaxis and treatment under a range of highly uncertain epidemiological parameters, this work provides public health decision-makers with an evidence-based guide for responding to a large-scale smallpox outbreak.
Collapse
|
22
|
Parker S, Crump R, Foster S, Hartzler H, Hembrador E, Lanier ER, Painter G, Schriewer J, Trost LC, Buller RM. Co-administration of the broad-spectrum antiviral, brincidofovir (CMX001), with smallpox vaccine does not compromise vaccine protection in mice challenged with ectromelia virus. Antiviral Res 2014; 111:42-52. [PMID: 25128688 PMCID: PMC9533899 DOI: 10.1016/j.antiviral.2014.08.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 07/31/2014] [Accepted: 08/04/2014] [Indexed: 12/02/2022]
Abstract
Natural orthopoxvirus outbreaks such as vaccinia, cowpox, cattlepox and buffalopox continue to cause morbidity in the human population. Monkeypox virus remains a significant agent of morbidity and mortality in Africa. Furthermore, monkeypox virus’s broad host-range and expanding environs make it of particular concern as an emerging human pathogen. Monkeypox virus and variola virus (the etiological agent of smallpox) are both potential agents of bioterrorism. The first line response to orthopoxvirus disease is through vaccination with first-generation and second-generation vaccines, such as Dryvax and ACAM2000. Although these vaccines provide excellent protection, their widespread use is impeded by the high level of adverse events associated with vaccination using live, attenuated virus. It is possible that vaccines could be used in combination with antiviral drugs to reduce the incidence and severity of vaccine-associated adverse events, or as a preventive in individuals with uncertain exposure status or contraindication to vaccination. We have used the intranasal mousepox (ectromelia) model to evaluate the efficacy of vaccination with Dryvax or ACAM2000 in conjunction with treatment using the broad spectrum antiviral, brincidofovir (BCV, CMX001). We found that co-treatment with BCV reduced the severity of vaccination-associated lesion development. Although the immune response to vaccination was quantifiably attenuated, vaccination combined with BCV treatment did not alter the development of full protective immunity, even when administered two days following ectromelia challenge. Studies with a non-replicating vaccine, ACAM3000 (MVA), confirmed that BCV’s mechanism of attenuating the immune response following vaccination with live virus was, as expected, by limiting viral replication and not through inhibition of the immune system. These studies suggest that, in the setting of post-exposure prophylaxis, co-administration of BCV with vaccination should be considered a first response to a smallpox emergency in subjects of uncertain exposure status or as a means of reduction of the incidence and severity of vaccine-associated adverse events.
Collapse
Affiliation(s)
- Scott Parker
- Saint Louis University School of Medicine, 1100 S. Grand Blvd., St. Louis, MO 63104, United States
| | - Ryan Crump
- Saint Louis University School of Medicine, 1100 S. Grand Blvd., St. Louis, MO 63104, United States
| | - Scott Foster
- Chimerix Inc., 2505 Meridian Parkway, Suite 340, Durham, NC 27713, United States
| | - Hollyce Hartzler
- Saint Louis University School of Medicine, 1100 S. Grand Blvd., St. Louis, MO 63104, United States
| | - Ed Hembrador
- Saint Louis University School of Medicine, 1100 S. Grand Blvd., St. Louis, MO 63104, United States
| | - E Randall Lanier
- Chimerix Inc., 2505 Meridian Parkway, Suite 340, Durham, NC 27713, United States
| | - George Painter
- Chimerix Inc., 2505 Meridian Parkway, Suite 340, Durham, NC 27713, United States
| | - Jill Schriewer
- Saint Louis University School of Medicine, 1100 S. Grand Blvd., St. Louis, MO 63104, United States
| | - Lawrence C Trost
- Chimerix Inc., 2505 Meridian Parkway, Suite 340, Durham, NC 27713, United States
| | - R Mark Buller
- Saint Louis University School of Medicine, 1100 S. Grand Blvd., St. Louis, MO 63104, United States.
| |
Collapse
|
23
|
Efficacy of tecovirimat (ST-246) in nonhuman primates infected with variola virus (Smallpox). Antimicrob Agents Chemother 2013; 57:6246-53. [PMID: 24100494 DOI: 10.1128/aac.00977-13] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Naturally occurring smallpox has been eradicated but remains a considerable threat as a biowarfare/bioterrorist weapon (F. Fleck, Bull. World Health Organ. 81:917-918, 2003). While effective, the smallpox vaccine is currently not recommended for routine use in the general public due to safety concerns (http://www.bt.cdc.gov/agent/smallpox/vaccination). Safe and effective countermeasures, particularly those effective after exposure to smallpox, are needed. Currently, SIGA Technologies is developing the small-molecule oral drug, tecovirimat (previously known as ST-246), as a postexposure therapeutic treatment of orthopoxvirus disease, including smallpox. Tecovirimat has been shown to be efficacious in preventing lethal orthopoxviral disease in numerous animal models (G. Yang, D. C. Pevear, M. H. Davies, M. S. Collett, T. Bailey, et al., J. Virol. 79:13139-13149, 2005; D. C. Quenelle, R. M. Buller, S. Parker, K. A. Keith, D. E. Hruby, et al., Antimicrob. Agents Chemother., 51:689-695, 2007; E. Sbrana, R. Jordan, D. E. Hruby, R. I. Mateo, S. Y. Xiao, et al., Am. J. Trop. Med. Hyg. 76:768-773, 2007). Furthermore, in clinical trials thus far, the drug appears to be safe, with a good pharmacokinetic profile. In this study, the efficacy of tecovirimat was evaluated in both a prelesional and postlesional setting in nonhuman primates challenged intravenously with 1 × 10(8) PFU of Variola virus (VARV; the causative agent of smallpox), a model for smallpox disease in humans. Following challenge, 50% of placebo-treated controls succumbed to infection, while all tecovirimat-treated animals survived regardless of whether treatment was started at 2 or 4 days postinfection. In addition, tecovirimat treatment resulted in dramatic reductions in dermal lesion counts, oropharyngeal virus shedding, and viral DNA circulating in the blood. Although clinical disease was evident in tecovirimat-treated animals, it was generally very mild and appeared to resolve earlier than in placebo-treated controls that survived infection. Tecovirimat appears to be an effective smallpox therapeutic in nonhuman primates, suggesting that it is reasonably likely to provide therapeutic benefit in smallpox-infected humans.
Collapse
|
24
|
Byrd CM, Grosenbach DW, Hruby DE. Antiviral options for biodefense. Curr Opin Virol 2013; 3:537-41. [DOI: 10.1016/j.coviro.2013.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 05/22/2013] [Accepted: 05/23/2013] [Indexed: 10/26/2022]
|
25
|
Smee DF. Orthopoxvirus inhibitors that are active in animal models: an update from 2008 to 2012. Future Virol 2013; 8:891-901. [PMID: 24563659 PMCID: PMC3929309 DOI: 10.2217/fvl.13.76] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antiviral agents are being sought as countermeasures for the potential deliberate release of smallpox (variola) and monkeypox viruses, for the treatment of naturally acquired monkeypox virus infections, and as therapy for complications due to smallpox (live-attenuated vaccinia virus) vaccination or accidental infection after exposure to vaccinated persons. Reviews of the scientific literature spanning 1950-2008 have documented the progress made in developing small-animal models of poxvirus infection and identifying novel antiviral agents. Compounds of considerable interest include cidofovir, CMX001 and ST-246® (tecovirimat; SIGA Technologies, NY, USA). New inhibitors have been identified since 2008, most of which do not exhibit the kind of potency and selectivity required for drug development. Two promising agents include 4'-thioidoxuridine (a nucleoside analog) and mDEF201 (an adenovirus-vectored interferon). Compounds that have been effectively used in combination studies include vaccinia immune globulin, cidofovir, ST-246 and CMX001. In the future there may be an increase in experimental work using active compounds in combination.
Collapse
Affiliation(s)
- Donald F Smee
- Institute for Antiviral Research, Department of Animal, Dairy & Veterinary Sciences, Utah State University, Logan, UT, 84322-5600, USA, Tel.: +1 435 797 2897, ,
| |
Collapse
|
26
|
De Clercq E. A cutting-edge view on the current state of antiviral drug development. Med Res Rev 2013; 33:1249-77. [PMID: 23495004 DOI: 10.1002/med.21281] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prominent in the current stage of antiviral drug development are: (i) for human immunodeficiency virus (HIV), the use of fixed-dose combinations (FDCs), the most recent example being Stribild(TM); (ii) for hepatitis C virus (HCV), the pleiade of direct-acting antivirals (DAAs) that should be formulated in the most appropriate combinations so as to obtain a cure of the infection; (iii)-(v) new strategies (i.e., AIC316, AIC246, and FV-100) for the treatment of herpesvirus infections: herpes simplex virus (HSV), cytomegalovirus (CMV), and varicella-zoster virus (VZV), respectively; (vi) the role of a new tenofovir prodrug, tenofovir alafenamide (TAF) (GS-7340) for the treatment of HIV infections; (vii) the potential use of poxvirus inhibitors (CMX001 and ST-246); (viii) the usefulness of new influenza virus inhibitors (peramivir and laninamivir octanoate); (ix) the position of the hepatitis B virus (HBV) inhibitors [lamivudine, adefovir dipivoxil, entecavir, telbivudine, and tenofovir disoproxil fumarate (TDF)]; and (x) the potential of new compounds such as FGI-103, FGI-104, FGI-106, dUY11, and LJ-001 for the treatment of filoviruses (i.e., Ebola). Whereas for HIV and HCV therapy is aimed at multiple-drug combinations, for all other viruses, HSV, CMV, VZV, pox, influenza, HBV, and filoviruses, current strategies are based on the use of single compounds.
Collapse
Affiliation(s)
- Erik De Clercq
- Rega Institute for Medical Research, KU Leuven, B-3000, Leuven, Belgium.
| |
Collapse
|
27
|
Effects of postchallenge administration of ST-246 on dissemination of IHD-J-Luc vaccinia virus in normal mice and in immune-deficient mice reconstituted with T cells. J Virol 2013; 87:5564-76. [PMID: 23468500 DOI: 10.1128/jvi.03426-12] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Whole-body bioimaging was used to study dissemination of vaccinia virus (VACV) in normal and in immune deficient (nu(-)/nu(-)) mice protected from lethality by postchallenge administration of ST-246. Total fluxes were recorded in the liver, spleen, lungs, and nasal cavities of live mice after intranasal infection with a recombinant IHD-J-Luc VACV expressing luciferase. Areas under the flux curve were calculated for individual mice to assess viral loads. Treatment for 2 to 5 days of normal BALB/c mice with ST-246 at 100 mg/kg starting 24 h postchallenge conferred 100% protection and reduced viral loads in four organs compared to control mice. Mice also survived after 5 days of treatment with ST-246 at 30 mg/kg, and yet the viral loads and poxes were higher in these mice compared to 100-mg/kg treatment group. Nude mice were not protected by ST-246 alone or by 10 million adoptively transferred T cells. In contrast, nude mice that received T cells and 7-day treatment with ST-246 survived infection and exhibited reduced viral loads compared to nonreconstituted and ST-246-treated mice after ST-246 was stopped. Similar protection of nude mice was achieved using adoptively transferred 1.0 and 0.1 million, but not 0.01 million, purified T cells or CD4(+) or CD8(+) T cells in conjunction with ST-246 treatment. These data suggest that ST-246 protects immunocompetent mice from lethality and reduces viral dissemination in internal organs and poxvirus lesions. Furthermore, immune-deficient animals with partial T cell reconstitution can control virus replication after a course of ST-246 and survive lethal vaccinia virus challenge.
Collapse
|
28
|
Finin P, Kosaraju A, Rose E, Rubin H. The Role of Vaccination, Antiorthopoxvirus Drug, and Social Cooperativity in a Mathematical Model of Smallpox Control. Biosecur Bioterror 2013; 11:59-72. [DOI: 10.1089/bsp.2012.0037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Peter Finin
- Peter Finin is a student at the University of Michigan Medical School, Ann Arbor, MI. Akhila Kosaraju, MD, is Vice President for Global Development, Siga Technologies, and Eric Rose, MD, is CEO of Siga Technologies, New York, NY. Dr. Rose is also Professor of Health Evidence and Policy, Mount Sinai School of Medicine, New York, NY. Harvey Rubin, MD, PhD, is Professor of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Akhila Kosaraju
- Peter Finin is a student at the University of Michigan Medical School, Ann Arbor, MI. Akhila Kosaraju, MD, is Vice President for Global Development, Siga Technologies, and Eric Rose, MD, is CEO of Siga Technologies, New York, NY. Dr. Rose is also Professor of Health Evidence and Policy, Mount Sinai School of Medicine, New York, NY. Harvey Rubin, MD, PhD, is Professor of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eric Rose
- Peter Finin is a student at the University of Michigan Medical School, Ann Arbor, MI. Akhila Kosaraju, MD, is Vice President for Global Development, Siga Technologies, and Eric Rose, MD, is CEO of Siga Technologies, New York, NY. Dr. Rose is also Professor of Health Evidence and Policy, Mount Sinai School of Medicine, New York, NY. Harvey Rubin, MD, PhD, is Professor of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Harvey Rubin
- Peter Finin is a student at the University of Michigan Medical School, Ann Arbor, MI. Akhila Kosaraju, MD, is Vice President for Global Development, Siga Technologies, and Eric Rose, MD, is CEO of Siga Technologies, New York, NY. Dr. Rose is also Professor of Health Evidence and Policy, Mount Sinai School of Medicine, New York, NY. Harvey Rubin, MD, PhD, is Professor of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
29
|
Protection of rabbits and immunodeficient mice against lethal poxvirus infections by human monoclonal antibodies. PLoS One 2012; 7:e48706. [PMID: 23133652 PMCID: PMC3487784 DOI: 10.1371/journal.pone.0048706] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 10/04/2012] [Indexed: 02/06/2023] Open
Abstract
Smallpox (variola virus) is a bioweapon concern. Monkeypox is a growing zoonotic poxvirus threat. These problems have resulted in extensive efforts to develop potential therapeutics that can prevent or treat potentially lethal poxvirus infections in humans. Monoclonal antibodies (mAbs) against smallpox are a conservative approach to this problem, as the licensed human smallpox vaccine (vaccinia virus, VACV) primarily works on the basis of protective antibody responses against smallpox. Fully human mAbs (hmAbs) against vaccinia H3 (H3L) and B5 (B5R), targeting both the mature virion (MV) and extracellular enveloped virion (EV) forms, have been developed as potential therapeutics for use in humans. Post-exposure prophylaxis was assessed in both murine and rabbit animal models. Therapeutic efficacy of the mAbs was assessed in three good laboratory practices (GLP) studies examining severe combined immunodeficiency mice (SCID) given a lethal VACV infection. Pre-exposure combination hmAb therapy provided significantly better protection against disease and death than either single hmAb or vaccinia immune globulin (VIG). Post-exposure combination mAb therapy provided significant protection against disease and death, and appeared to fully cure the VACV infection in ≥50% of SCID mice. Therapeutic efficacy was then assessed in two rabbit studies examining post-exposure hmAb prophylaxis against rabbitpox (RPXV). In the first study, rabbits were infected with RPVX and then provided hmAbs at 48 hrs post-infection, or 1 hr and 72 hrs post-infection. Rabbits in both groups receiving hmAbs were 100% protected from death. In the second rabbitpox study, 100% of animal treated with combination hmAb therapy and 100% of animals treated with anti-B5 hmAb were protected. These findings suggest that combination hmAb treatment may be effective at controlling smallpox disease in immunocompetent or immunodeficient humans.
Collapse
|
30
|
Characterization of ectromelia virus deficient in EVM036, the homolog of vaccinia virus F13L, and its application for rapid generation of recombinant viruses. J Virol 2012; 86:13501-7. [PMID: 23035222 DOI: 10.1128/jvi.01732-12] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The orthopoxvirus (OPV) vaccinia virus (VACV) requires an intact F13L gene to produce enveloped virions (EV) and to form plaques in cell monolayers. Simultaneous introduction of an exogenous gene and F13L into F13L-deficient VACV results in expression of the foreign gene and restoration of plaque size. This is used as a method to rapidly generate VACV recombinants without the need for drug selection. However, whether other OPVs require the orthologs of F13L to generate EV and form plaques, whether F13L orthologs and EV are important for OPV pathogenesis in natural hosts, and whether a system based on F13L ortholog deficiency can be used to generate recombinant OPVs other than VACV have not been reported. The F13L ortholog in ectromelia virus (ECTV), the agent of mousepox, is EVM036. We show that ECTV lacking EVM036 formed small plaques and was highly attenuated in vivo but still induced strong antibody responses. Reintroduction of EVM036 in tandem with the DsRed gene resulted in a virus that expressed DsRed in infected cells but was indistinguishable from wild-type ECTV in terms of plaque size and in vivo virulence. Thus, our data show that, like F13L in VACV, EVM036 is required for ECTV plaque formation and that EVM036 and EV are important for ECTV virulence. Our experiments also suggest that OPVs deficient in F13L orthologs could serve as safer anti-OPV vaccines. Further, our results demonstrate that ECTV deficient in EVM036 can be exploited for the rapid generation of fully virulent ECTV expressing foreign genes of interest.
Collapse
|
31
|
De Clercq E. Human viral diseases: what is next for antiviral drug discovery? Curr Opin Virol 2012; 2:572-9. [PMID: 22846888 DOI: 10.1016/j.coviro.2012.07.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 07/10/2012] [Indexed: 01/30/2023]
Abstract
For the treatment of human immunodeficiency virus (HIV) infections for which there are ample drugs available, the immediate future lies in a once-daily combination pill containing three or four active ingredients. This strategy may also be envisaged for the treatment of hepatitis C virus (HCV) infections as soon as we have at hand the appropriate direct-acting antiviral agents (DAAs) to be combined. A combination drug therapy is generally not entertained for other viruses. Yet, new drugs are at the horizon for the treatment of herpes simplex virus (HSV), varicella-zoster virus (VZV), poxvirus, hepatitis B virus (HBV), influenza and enveloped viruses-at-large.
Collapse
Affiliation(s)
- Erik De Clercq
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium.
| |
Collapse
|
32
|
Israely T, Paran N, Lustig S, Erez N, Politi B, Shafferman A, Melamed S. A single cidofovir treatment rescues animals at progressive stages of lethal orthopoxvirus disease. Virol J 2012; 9:119. [PMID: 22709563 PMCID: PMC3409050 DOI: 10.1186/1743-422x-9-119] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 06/18/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In an event of a smallpox outbreak in humans, the window for efficacious treatment by vaccination with vaccinia viruses (VACV) is believed to be limited to the first few days post-exposure (p.e.). We recently demonstrated in a mouse model for human smallpox, that active immunization 2-3 days p.e. with either VACV-Lister or modified VACV Ankara (MVA) vaccines, can rescue animals from lethal challenge of ectromelia virus (ECTV), the causative agent of mousepox. The present study was carried out in order to determine whether a single dose of the anti-viral cidofovir (CDV), administered at different times and doses p.e. either alone or in conjunction with active vaccination, can rescue ECTV infected mice. METHODS Animals were infected intranasally with ECTV, treated on different days with various single CDV doses and monitored for morbidity, mortality and humoral response. In addition, in order to determine the influence of CDV on the immune response following vaccination, both the "clinical take", IFN-gamma and IgG Ab levels in the serum were evaluated as well as the ability of the mice to withstand a lethal challenge of ECTV. Finally the efficacy of a combined treatment regime of CDV and vaccination p.e. was determined. RESULTS A single p.e. CDV treatment is sufficient for protection depending on the initiation time and dose (2.5 - 100 mg/kg) of treatment. Solid protection was achieved by a low dose (5 mg/kg) CDV treatment even if given at day 6 p.e., approximately 4 days before death of the control infected untreated mice (mean time to death (MTTD) 10.2). At the same time point complete protection was achieved by single treatment with higher doses of CDV (25 or 100 mg/kg). Irrespective of treatment dose, all surviving animals developed a protective immune response even when the CDV treatment was initiated one day p.e.. After seven days post treatment with the highest dose (100 mg/kg), virus was still detected in some organs (e.g. lung and liver) yet all animals survived, suggesting that efficacious single CDV treatment requires a potent immune system. The combination of CDV and vaccination provided no additional protection over CDV alone. Yet, combining CDV and vaccination maintained vaccination efficacy. CONCLUSIONS Altogether, our data substantiate the feasibility of single post-exposure antiviral treatment to face orthopoxvirus infection.
Collapse
Affiliation(s)
- Tomer Israely
- Israel Institute for Biological Research, Ness-ziona, Israel
| | | | | | | | | | | | | |
Collapse
|
33
|
Selvy PE, Lavieri RR, Lindsley CW, Brown HA. Phospholipase D: enzymology, functionality, and chemical modulation. Chem Rev 2011; 111:6064-119. [PMID: 21936578 PMCID: PMC3233269 DOI: 10.1021/cr200296t] [Citation(s) in RCA: 280] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Paige E Selvy
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37064, USA
| | | | | | | |
Collapse
|
34
|
Kroon EG, Mota BEF, Abrahão JS, da Fonseca FG, de Souza Trindade G. Zoonotic Brazilian Vaccinia virus: from field to therapy. Antiviral Res 2011; 92:150-63. [PMID: 21896287 DOI: 10.1016/j.antiviral.2011.08.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 08/19/2011] [Accepted: 08/22/2011] [Indexed: 12/27/2022]
Abstract
Vaccinia virus (VACV), the prototype species of the Orthopoxvirus (OPV) genus, causes an occupational zoonotic disease in Brazil that is primarily associated with the handling of infected dairy cattle. Cattle and human outbreaks have been described in southeastern Brazil since 1999 and have now occurred in almost half of the territory. Phylogenetic studies have shown high levels of polymorphisms among isolated VACVs, which indicate the existence of at least two genetically divergent clades; this has also been proven in virulence assays in a mouse model system. In humans, VACV infection is characterized by skin lesions, primarily on the hands, accompanied by systemic symptoms such as fever, myalgia, headache and lymphadenopathy. In this review, we will discuss the virological, epidemiological, ecological and clinical aspects of VACV infection, its diagnosis and compounds that potentially could be used for the treatment of severe cases.
Collapse
Affiliation(s)
- Erna Geessien Kroon
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG 31270-901, Brazil.
| | | | | | | | | |
Collapse
|
35
|
Tomimori Y, Kawakami Y, McCausland MM, Ando T, Koriazova L, Kato S, Kawakami T, Crotty S. Protective murine and human monoclonal antibodies against eczema vaccinatum. Antivir Ther 2011; 16:67-75. [PMID: 21311110 DOI: 10.3851/imp1717] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Eczema vaccinatum is the most common severe pathology associated with smallpox vaccination (vaccinia virus), occurring at high rates among individuals with a previous history of atopic dermatitis (atopic eczema). METHODS Monoclonal antibodies capable of neutralizing vaccinia virus, anti-H3 and anti-B5, were developed as a potential therapy for treatment of human eczema vaccinatum. RESULTS Using a small animal model of eczema vaccinatum, we demonstrated that both murine and fully human monoclonal antibodies effectively limited eczema vaccinatum disease, foreshortening both the disease kinetics and the severity of the erosive viral skin lesions. CONCLUSIONS These neutralizing antibodies would likely be effective at reducing or eliminating clinical disease in people with eczema vaccinatum or other severe side effects of the smallpox vaccine.
Collapse
Affiliation(s)
- Yoshiaki Tomimori
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology (LIAI), La Jolla, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Duraffour S, Matthys P, van den Oord JJ, De Schutter T, Mitera T, Snoeck R, Andrei G. Study of camelpox virus pathogenesis in athymic nude mice. PLoS One 2011; 6:e21561. [PMID: 21738709 PMCID: PMC3125194 DOI: 10.1371/journal.pone.0021561] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 06/02/2011] [Indexed: 11/29/2022] Open
Abstract
Camelpox virus (CMLV) is the closest known orthopoxvirus genetically related to variola virus. So far, CMLV was restricted to camelids but, recently, three human cases of camelpox have been described in India, highlighting the need to pursue research on its pathogenesis, which has been hampered by the lack of small animal models. Here, we confirm that NMRI immunocompetent mice are resistant to intranasal (i.n.) CMLV infection. However, we demonstrate that CMLV induced a severe disease following i.n. challenge of athymic nude mice, which was accompanied with a failure in gaining weight, leading to euthanasia of the animals. On the other hand, intracutaneous (i.c.) infection resulted in disease development without impacting the body weight evolution. CMLV replication in tissues and body fluids was confirmed in the two models. We further analyzed innate immune and B cell responses induced in the spleen and draining lymph nodes after exposure to CMLV. In both models, strong increases in CD11b+F4/80+ macrophages were seen in the spleen, while neutrophils, NK and B cell responses varied between the routes of infection. In the lymph nodes, the magnitude of CD11c+CD8α+ lymphoid and CD11c+CD11b+ myeloid dendritic cell responses increased in i.n. challenged animals. Analysis of cytokine profiles revealed significant increases of interleukin (IL)-6 and IL-18 in the sera of infected animals, while those of other cytokines were similar to uninfected controls. The efficacy of two antivirals (cidofovir or HPMPC, and its 2, 6-diaminopurine analog) was evaluated in both models. HPMPC was the most effective molecule affording 100% protection from morbidity. It appeared that both treatments did not affect immune cell responses or cytokine expression. In conclusion, we demonstrated that immunodeficient mice are permissive for CMLV propagation. These results provide a basis for studying the pathogenesis of CMLV, as well as for evaluating potential antiviral therapies in an immunodeficiency context.
Collapse
Affiliation(s)
- Sophie Duraffour
- Rega Institute, Laboratory of Virology and Chemotherapy, K.U.L, Leuven, Belgium.
| | | | | | | | | | | | | |
Collapse
|
37
|
Grosenbach DW, Jordan R, Hruby DE. Development of the small-molecule antiviral ST-246 as a smallpox therapeutic. Future Virol 2011; 6:653-671. [PMID: 21837250 DOI: 10.2217/fvl.11.27] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Naturally occurring smallpox has been eradicated, yet it remains as one of the highest priority pathogens due to its potential as a biological weapon. The majority of the US population would be vulnerable in a smallpox outbreak. SIGA Technologies, Inc. has responded to the call of the US government to develop and supply to the Strategic National Stockpile a smallpox antiviral to be deployed in the event of a smallpox outbreak. ST-246(®) (tecovirimat) was initially identified via a high-throughput screen in 2002, and in the ensuing years, our drug-development activities have spanned in vitro analysis, preclinical safety, pharmacokinetics and efficacy testing (all according to the 'animal rule'). Additionally, SIGA has conducted Phase I and II clinical trials to evaluate the safety, tolerability and pharmacokinetics of ST-246, bringing us to our current late stage of clinical development. This article reviews the need for a smallpox therapeutic and our experience in developing ST-246, and provides perspective on the role of a smallpox antiviral during a smallpox public health emergency.
Collapse
Affiliation(s)
- Douglas W Grosenbach
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230, Corvallis, OR 97333, USA
| | | | | |
Collapse
|
38
|
Torres E, Duque MD, Camps P, Naesens L, Calvet T, Font-Bardia M, Vázquez S. Polycyclic N-benzamido imides with potent activity against vaccinia virus. ChemMedChem 2011; 5:2072-8. [PMID: 20967819 PMCID: PMC7162373 DOI: 10.1002/cmdc.201000306] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The synthesis and antiviral activity of a series of novel polycyclic analogues of the orthopoxvirus egress inhibitor tecovirimat (ST-246) is presented. Several of these compounds display sub-micromolar activity against vaccinia virus, and were more potent than cidofovir (CDV). The more active compounds were about 10-fold more active than CDV, with minimum cytotoxic concentrations above 100 μM. Chemical manipulations of the two carbon-carbon double bonds present in the compounds were carried out to further explore the structure-activity relationships of these new polycyclic imides. Hydrogenation of the two carbon-carbon double bonds decreases antiviral activity, whereas either cyclopropanation or epoxidation of the double bonds fully eliminates the antiviral activity.
Collapse
Affiliation(s)
- Eva Torres
- Institute of Biomedicine, Universitat de Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
39
|
Treatment of Vaccinia and Cowpox Virus Infections in Mice with CMX001 and ST-246. Viruses 2010; 2:2681-95. [PMID: 21994637 PMCID: PMC3185598 DOI: 10.3390/v2122681] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 12/06/2010] [Accepted: 12/06/2010] [Indexed: 11/26/2022] Open
Abstract
Although a large number of compounds have been identified with antiviral activity against orthopoxviruses in tissue culture systems, it is highly preferred that these compounds have activity in vivo before they can be seriously considered for further development. One of the most commonly used animal models for the confirmation of this activity has been the use of mice infected with either vaccinia or cowpox viruses. These model systems have the advantage that they are relatively inexpensive, readily available and do not require any special containment facilities; therefore, relatively large numbers of compounds can be evaluated in vivo for their activity. The two antiviral agents that have progressed from preclinical studies to human safety trials for the treatment of orthopoxvirus infections are the cidofovir analog, CMX001, and an inhibitor of extracellular virus formation, ST-246. These compounds are the ones most likely to be used in the event of a bioterror attack. The purpose of this communication is to review the advantages and disadvantages of using mice infected with vaccinia and cowpox virus as surrogate models for human orthopoxvirus infections and to summarize the activity of CMX001 and ST-246 in these model infections.
Collapse
|
40
|
Chen N, Bellone CJ, Schriewer J, Owens G, Fredrickson T, Parker S, Buller RML. Poxvirus interleukin-4 expression overcomes inherent resistance and vaccine-induced immunity: pathogenesis, prophylaxis, and antiviral therapy. Virology 2010; 409:328-37. [PMID: 21071055 DOI: 10.1016/j.virol.2010.10.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 09/14/2010] [Accepted: 10/12/2010] [Indexed: 10/18/2022]
Abstract
In 2001, Jackson et al. reported that murine IL-4 expression by a recombinant ectromelia virus caused enhanced morbidity and lethality in resistant C57BL/6 mice as well as overcame protective immune memory responses. To achieve a more thorough understanding of this phenomenon and to assess a variety of countermeasures, we constructed a series of ECTV recombinants encoding murine IL-4 under the control of promoters of different strengths and temporal regulation. We showed that the ECTV-IL-4 recombinant expressing the highest level of IL-4 was uniformly lethal for C57BL/6 mice even when previously immunized. The lethality of the ECTV-IL-4 recombinants resulted from virus-expressed IL-4 signaling through the IL-4 receptor but was not due to IL-4 toxicity. A number of treatment approaches were evaluated against the most virulent IL-4 encoding virus. The most efficacious therapy was a combination of two antiviral drugs (CMX001(®) and ST-246(®)) that have different mechanisms of action.
Collapse
Affiliation(s)
- Nanhai Chen
- Genelux Corporation, San Diego Science Center, 3030 Bunker Hill Street, Suite 310, San Diego, CA 92109, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Dropulic LK, Cohen JI. Update on new antivirals under development for the treatment of double-stranded DNA virus infections. Clin Pharmacol Ther 2010; 88:610-9. [PMID: 20881959 PMCID: PMC3426500 DOI: 10.1038/clpt.2010.178] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
All the currently available antiviral agents used in the treatment of double-stranded (ds) DNA viruses, with the exception of interferon-α, inhibit the same target, the viral DNA polymerase. With increasing reports of the development of resistance of herpes simplex virus (HSV), cytomegalovirus (CMV), and hepatitis B virus (HBV) to some of these drugs, new antiviral agents are needed to treat these infections. Additionally, no drugs have been approved to treat several DNA virus infections, including those caused by adenovirus, smallpox, molluscum contagiosum, and BK virus. We report the status of 10 new antiviral drugs for the treatment of dsDNA viruses. CMX-001 has broad activity against dsDNA viruses; 3 helicase-primase inhibitors, maribavir, and FV-100 have activity against certain herpesviruses; ST-246 inhibits poxviruses; GS-9191 inhibits papillomaviruses; and clevudine and emtricitabine are active against HBV. Most of these drugs have completed at least phase I trials in humans, and many are in additional clinical trials.
Collapse
Affiliation(s)
- Lesia K. Dropulic
- Medical Virology Section, Laboratory of Clinical Infectious Diseases, Bldg. 10, Room 11N234, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892
| | - Jeffrey I. Cohen
- Medical Virology Section, Laboratory of Clinical Infectious Diseases, Bldg. 10, Room 11N234, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892
| |
Collapse
|
42
|
Jordan R, Leeds JM, Tyavanagimatt S, Hruby DE. Development of ST-246® for Treatment of Poxvirus Infections. Viruses 2010; 2:2409-2435. [PMID: 21994624 PMCID: PMC3185582 DOI: 10.3390/v2112409] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Revised: 10/26/2010] [Accepted: 10/26/2010] [Indexed: 12/26/2022] Open
Abstract
ST-246 (Tecovirimat) is a small synthetic antiviral compound being developed to treat pathogenic orthopoxvirus infections of humans. The compound was discovered as part of a high throughput screen designed to identify inhibitors of vaccinia virus-induced cytopathic effects. The antiviral activity is specific for orthopoxviruses and the compound does not inhibit the replication of other RNA- and DNA-containing viruses or inhibit cell proliferation at concentrations of compound that are antiviral. ST-246 targets vaccinia virus p37, a viral protein required for envelopment and secretion of extracellular forms of virus. The compound is orally bioavailable and protects multiple animal species from lethal orthopoxvirus challenge. Preclinical safety pharmacology studies in mice and non-human primates indicate that ST-246 is readily absorbed by the oral route and well tolerated with the no observable adverse effect level (NOAEL) in mice measured at 2000 mg/kg and the no observable effect level (NOEL) in non-human primates measured at 300 mg/kg. Drug substance and drug product processes have been developed and commercial scale batches have been produced using Good Manufacturing Processes (GMP). Human phase I clinical trials have shown that ST-246 is safe and well tolerated in healthy human volunteers. Based on the results of the clinical evaluation, once a day dosing should provide plasma drug exposure in the range predicted to be antiviral based on data from efficacy studies in animal models of orthopoxvirus disease. These data support the use of ST-246 as a therapeutic to treat pathogenic orthopoxvirus infections of humans.
Collapse
Affiliation(s)
- Robert Jordan
- SIGA Technologies, 4575 SW Research Way, Corvallis, OR 97333, USA; E-Mails: (J.M.L); (S.T.); (D.E.H.)
| | - Janet M. Leeds
- SIGA Technologies, 4575 SW Research Way, Corvallis, OR 97333, USA; E-Mails: (J.M.L); (S.T.); (D.E.H.)
| | | | - Dennis E. Hruby
- SIGA Technologies, 4575 SW Research Way, Corvallis, OR 97333, USA; E-Mails: (J.M.L); (S.T.); (D.E.H.)
| |
Collapse
|
43
|
Berhanu A, King DS, Mosier S, Jordan R, Jones KF, Hruby DE, Grosenbach DW. Impact of ST-246® on ACAM2000™ smallpox vaccine reactogenicity, immunogenicity, and protective efficacy in immunodeficient mice. Vaccine 2010; 29:289-303. [PMID: 21036130 DOI: 10.1016/j.vaccine.2010.10.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 09/29/2010] [Accepted: 10/13/2010] [Indexed: 11/16/2022]
Abstract
Although a highly effective vaccine against smallpox, vaccinia virus (VV) is not without adverse events, some of which can be life-threatening, particularly in immunocompromised individuals. We have recently demonstrated that the immunogenicity and protective efficacy of Dryvax(®) in immunocompetent mice is preserved even when co-administered with ST-246, an orally bioavailable small-molecule inhibitor of orthopoxvirus egress and dissemination. In addition, ST-246 markedly reduced the reactogenicity of the smallpox vaccine ACAM2000 and the highly neurovirulent VV strain Western Reserve (VV-WR). Here, we evaluated the impact of ST-246 co-administration on ACAM2000 reactogenicity, immunogenicity, and protective efficacy in seven murine models of varying degrees of humoral and cellular immunodeficiency: BALB/c and B-cell deficient (JH-KO) mice depleted of CD4(+) or CD8(+) or both subsets of T cells. We observed that ST-246 reduced vaccine lesion severity and time to complete resolution in all of the immunodeficient models examined, except in those lacking both CD4(+) and CD8(+) T cells. Although VV-specific humoral responses were moderately reduced by ST-246 treatment, cellular responses were generally comparable or slightly enhanced at both 1 and 6 months post-vaccination. Most importantly, in those models in which vaccination given alone conferred protection against lethal VV challenge, similar levels of protection were observed at both time points when vaccination was given with ST-246. These data suggest that, with the exception of individuals with irreversible, combined CD4(+) and CD8(+) T-cell deficiency, ST-246 co-administered at the time of vaccination may help reduce vaccine reactogenicity--even in those lacking humoral immunity--without impeding the induction of protective immunity.
Collapse
Affiliation(s)
- Aklile Berhanu
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230, Corvallis, OR 97333, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Parker S, Siddiqui AM, Painter G, Schriewer J, Buller RM. Ectromelia virus infections of mice as a model to support the licensure of anti-orthopoxvirus therapeutics. Viruses 2010; 2:1918-1932. [PMID: 21994714 PMCID: PMC3185751 DOI: 10.3390/v2091918] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 08/30/2010] [Accepted: 08/31/2010] [Indexed: 12/02/2022] Open
Abstract
The absence of herd immunity to orthopoxviruses and the concern that variola or monkeypox viruses could be used for bioterroristic activities has stimulated the development of therapeutics and safer prophylactics. One major limitation in this process is the lack of accessible human orthopoxvirus infections for clinical efficacy trials; however, drug licensure can be based on orthopoxvirus animal challenge models as described in the "Animal Efficacy Rule". One such challenge model uses ectromelia virus, an orthopoxvirus, whose natural host is the mouse and is the etiological agent of mousepox. The genetic similarity of ectromelia virus to variola and monkeypox viruses, the common features of the resulting disease, and the convenience of the mouse as a laboratory animal underscores its utility in the study of orthopoxvirus pathogenesis and in the development of therapeutics and prophylactics. In this review we outline how mousepox has been used as a model for smallpox. We also discuss mousepox in the context of mouse strain, route of infection, infectious dose, disease progression, and recovery from infection.
Collapse
Affiliation(s)
- Scott Parker
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, 1100 S. Grand Blvd., St. Louis, MO, 63104, USA; E-Mails: (S.P.); (A.M.S.); (J.S.)
| | - Akbar M. Siddiqui
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, 1100 S. Grand Blvd., St. Louis, MO, 63104, USA; E-Mails: (S.P.); (A.M.S.); (J.S.)
| | - George Painter
- Chimerix Inc., 2505 Meridian Park Way, Suite 340, Durham, NC, 27713, USA; E-Mail:
| | - Jill Schriewer
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, 1100 S. Grand Blvd., St. Louis, MO, 63104, USA; E-Mails: (S.P.); (A.M.S.); (J.S.)
| | - R. Mark Buller
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, 1100 S. Grand Blvd., St. Louis, MO, 63104, USA; E-Mails: (S.P.); (A.M.S.); (J.S.)
| |
Collapse
|