1
|
Tamir H, Noy-Porat T, Melamed S, Cherry-Mimran L, Barlev-Gross M, Alcalay R, Yahalom-Ronen Y, Achdout H, Politi B, Erez N, Weiss S, Rosenfeld R, Epstein E, Mazor O, Makdasi E, Paran N, Israely T. Synergistic effect of two human-like monoclonal antibodies confers protection against orthopoxvirus infection. Nat Commun 2024; 15:3265. [PMID: 38627363 PMCID: PMC11021552 DOI: 10.1038/s41467-024-47328-y] [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: 09/14/2023] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
The eradication of smallpox was officially declared by the WHO in 1980, leading to discontinuation of the vaccination campaign against the virus. Consequently, immunity against smallpox and related orthopoxviruses like Monkeypox virus gradually declines, highlighting the need for efficient countermeasures not only for the prevention, but also for the treatment of already exposed individuals. We have recently developed human-like monoclonal antibodies (mAbs) from vaccinia virus-immunized non-human primates. Two mAbs, MV33 and EV42, targeting the two infectious forms of the virus, were selected for in vivo evaluation, based on their in vitro neutralization potency. A single dose of either MV33 or EV42 administered three days post-infection (dpi) to BALB/c female mice provides full protection against lethal ectromelia virus challenge. Importantly, a combination of both mAbs confers full protection even when provided five dpi. Whole-body bioimaging and viral load analysis reveal that combination of the two mAbs allows for faster and more efficient clearance of the virus from target organs compared to either MV33 or EV42 separately. The combined mAbs treatment further confers post-exposure protection against the currently circulating Monkeypox virus in Cast/EiJ female mice, highlighting their therapeutic potential against other orthopoxviruses.
Collapse
Affiliation(s)
- Hadas Tamir
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Tal Noy-Porat
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Sharon Melamed
- Israel Institute for Biological Research, Ness Ziona, Israel
| | | | | | - Ron Alcalay
- Israel Institute for Biological Research, Ness Ziona, Israel
| | | | - Hagit Achdout
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Boaz Politi
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Noam Erez
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shay Weiss
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Ronit Rosenfeld
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Eyal Epstein
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Ohad Mazor
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Efi Makdasi
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Nir Paran
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Tomer Israely
- Israel Institute for Biological Research, Ness Ziona, Israel.
| |
Collapse
|
2
|
Nucera F, Bonina L, Cipolla A, Pirina P, Hansbro PM, Adcock IM, Caramori G. Poxviridae Pneumonia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:183-204. [PMID: 38801579 DOI: 10.1007/978-3-031-57165-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Poxviridae family includes several viruses that infecting humans usually causes skin lesions only, but in some cases their clinical course is complicated by viral pneumonia (with or without bacterial superinfections). Historically variola virus has been the poxviridae most frequently associated with the development of pneumonia with many large outbreaks worldwide before its eradication in 1980. It is still considered a biological threat for its potential in biological warfare and bioterrorism. Smallpox pneumonia can be severe with the onset of acute respiratory distress syndrome (ARDS) and death. Vaccinia virus, used for vaccination against smallpox exceptionally, in immunocompromised patients, can induce generalized (with also lung involvement) severe disease after vaccination. MPXV virus occasionally can cause pneumonia particularly in immunocompromised patients. The pathophysiology of poxviridae pneumonia is still an area of active research; however, in animal models these viruses can cause both direct damage to the lower airways epithelium and a hyperinflammatory syndrome, like a cytokine storm. Multiple mechanisms of immune evasion have also been described. The treatment of poxviridae pneumonia is mainly based on careful supportive care. Despite the absence of randomized clinical trials in patients with poxviridae pneumonia there are antiviral drugs, such as tecovirimat, cidofovir and brincidofovir, FDA-approved for use in smallpox and also available under an expanded access protocol for treatment of MPXV. There are 2 (replication-deficient modified vaccinia Ankara and replication-competent vaccinia virus) smallpox vaccines FDA-approved with the first one also approved for prevention of MPXV in adults that are at high risk of infection.
Collapse
Affiliation(s)
- Francesco Nucera
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Letterio Bonina
- Virologia, Dipartimento di Patologia delle Malattie Umane "G. Barresi", Università degli Studi di Messina, Messina, Italy
| | - Antonino Cipolla
- Pneumologia, Dipartimento di Medicina Clinica e Sperimentale, Università degli Studi di Catania, Catania, Italy
| | - Pietro Pirina
- Pneumologia, Dipartimento di Medicina, Chirurgia e Farmacia, Università degli Studi di Sassari, Sassari, Italy
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Gaetano Caramori
- Pulmonology, Department of Medicine and Surgery, University of Parma, Parma, Italy.
| |
Collapse
|
3
|
Asadi Noghabi F, G. Rizk J, Makkar D, Roozbeh N, Ghelichpour S, Zarei A. Managing Monkeypox Virus Infections: A Contemporary Review. IRANIAN JOURNAL OF MEDICAL SCIENCES 2024; 49:1-9. [PMID: 38322157 PMCID: PMC10839137 DOI: 10.30476/ijms.2022.96738.2837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/30/2022] [Accepted: 11/22/2022] [Indexed: 02/08/2024]
Abstract
Monkeypox is an infectious and contagious zoonotic disease caused by the Orthopoxvirus species and was first identified in Africa. Recently, this infectious disease has spread widely in many parts of the world. Fever, fatigue, headache, and rash are common symptoms of monkeypox. The presence of lymphadenopathy is another prominent and key symptom of monkeypox, which distinguishes this disease from other diseases and is useful for diagnosing the disease. This disease is transmitted to humans through contact with or eating infected animals as well as objects infected with the virus. One of the ways to diagnose this disease is through PCR testing of lesions and secretions. To prevent the disease, vaccines such as JYNNEOS and ACAM2000 are available, but they are not accessible to all people in the world, and their effectiveness and safety need further investigation. However, preventive measures such as avoiding contact with people infected with the virus and using appropriate personal protective equipment are mandatory. The disease therapy is based on medicines such as brincidofovir, cidofovir, and Vaccinia Immune Globulin Intravenous. The injectable format of tecovirimat was approved recently, in May 2022. Considering the importance of clinical care in this disease, awareness about the side effects of medicines, nutrition, care for conjunctivitis, skin rash, washing and bathing at home, and so on can be useful in controlling and managing the disease.
Collapse
Affiliation(s)
- Fariba Asadi Noghabi
- Department of Nursing, School of Nursing and Midwifery, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - John G. Rizk
- Department of Pharmaceutical Health Services Research Center, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | | | - Nasibeh Roozbeh
- Mother and Child Welfare Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Soleyman Ghelichpour
- Student Research Committee, School of Nursing and Midwifery, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Aref Zarei
- Department of Nursing, School of Nursing and Midwifery, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
4
|
Monkeypox infection elicits strong antibody and B cell response against A35R and H3L antigens. iScience 2023; 26:105957. [PMID: 36687315 PMCID: PMC9838220 DOI: 10.1016/j.isci.2023.105957] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/19/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
Monkeypox virus (MPXV) resides in two forms; mature and enveloped, and depending on it, distinct proteins are displayed on the viral surface. Here, we expressed two MPXV antigens from the mature, and one from the enveloped form, and tested their reactivity to sera of 11 MPXV recoverees while comparing to sera from recently and past vaccinated individuals. 8 out of 11 recoverees exhibited detectable neutralization levels against Vaccinia Lister. Sera from all recoverees bound strongly to A35R and H3L antigens. Moreover, the responses to A35R were significantly higher within the recoverees compared to both recently and past vaccinated donors. Lastly, A35R- and H3L-specific IgG+ B cells ranging from 0.03-0.46% and 0.11-0.36%, respectively, were detected in all recoverees (A35R), and in 9 out of 11 recoverees (H3L). Therefore, A35R and H3L represent MPXV immune targets and could be used in a heat-inactivated serological ELISA for the identification of recent MPXV infection.
Collapse
|
5
|
Aldhaeefi M, Rungkitwattanakul D, Unonu J, Franklin CJ, Lyons J, Hager K, Daftary MN. The 2022 human monkeypox outbreak: Clinical review and management guidance. Am J Health Syst Pharm 2023; 80:44-52. [PMID: 36259674 DOI: 10.1093/ajhp/zxac300] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Indexed: 01/19/2023] Open
Abstract
PURPOSE The objective of this clinical review is to provide an overview of antiviral therapies for monkeypox treatment and summarize the role of vaccines in monkeypox prevention. SUMMARY The human monkeypox virus is a double-stranded DNA virus of the Orthopoxvirus genus of the Poxviridae family. The estimated case fatality rate for monkeypox ranges between 0% and 11%. The first human monkeypox infection was reportedly due to an unidentified animal reservoir. Per the Centers for Disease Control and Prevention, isolation and infection control procedures should be followed in the care of those infected with monkeypox virus. Monkeypox virus infection symptoms include rash, fever, chills, headache, muscle aches, backache, and fatigue that may progress to exhaustion. Severe complications such as encephalitis, pneumonia, and retropharyngeal abscess could appear in immunocompromised or critically ill patients. There are currently no specific Food and Drug Administration (FDA)-approved therapies for monkeypox. As with most viral infections, supportive care is the backbone of monkeypox clinical management. However, therapies effective for smallpox, such as cidofovir, brincidofovir, and tecovirimat, have previously been reported to be effective in the management of monkeypox. Pre- and postexposure prophylaxis to prevent monkeypox transmission are recommended in the US for those at high risk for disease transmission. CONCLUSION There are no FDA-approved treatments for monkeypox infection. Surveillance and detection of monkeypox among high-risk populations should be implemented to help understand the epidemiology of this disease.
Collapse
Affiliation(s)
- Mohammed Aldhaeefi
- Department of Clinical and Administrative Pharmacy Sciences, College of Pharmacy, Howard University, Washington, DC, USA
| | - Dhakrit Rungkitwattanakul
- Department of Clinical and Administrative Pharmacy Sciences, College of Pharmacy, Howard University, Washington, DC, USA
| | - Jacqueise Unonu
- Department of Clinical and Administrative Pharmacy Sciences, College of Pharmacy, Howard University, Washington, DC, USA
| | - Careen-Joan Franklin
- Department of Clinical and Administrative Pharmacy Sciences, College of Pharmacy, Howard University, Washington, DC, USA
| | - Jessica Lyons
- Department of Clinical and Administrative Pharmacy Sciences, College of Pharmacy, Howard University, Washington, DC, USA
| | - Katherine Hager
- Division of Infectious Diseases, Department of Medicine, Howard University College of Medicine, Washington, DC, USA
| | - Monika N Daftary
- Department of Clinical and Administrative Pharmacy Sciences, College of Pharmacy, Howard University, Washington, DC, USA
| |
Collapse
|
6
|
Therapeutic strategies for human poxvirus infections: Monkeypox (mpox), smallpox, molluscipox, and orf. Travel Med Infect Dis 2022; 52:102528. [PMID: 36539022 PMCID: PMC9758798 DOI: 10.1016/j.tmaid.2022.102528] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/01/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Therapeutic and vaccine development for human poxvirus infections (e.g., monkeypox (mpox) virus, variola virus, molluscum contagiosum virus, orf virus) has been largely deserted, especially after the eradication of smallpox by 1980. Human mpox is a self-limited disease confined to Central and West Africa for decades. However, since April 2022, mpox has quickly emerged as a multi-country outbreak, urgently calling for effective antiviral agents and vaccines to control mpox. Here, this review highlights possible therapeutic options (e.g., tecovirimat, brincidofovir, cidofovir) and other strategies (e.g., vaccines, intravenous vaccinia immune globulin) for the management of human poxvirus infections worldwide.
Collapse
|
7
|
Islam MR, Hossain MJ, Roy A, Hasan AHMN, Rahman MA, Shahriar M, Bhuiyan MA. Repositioning potentials of smallpox vaccines and antiviral agents in monkeypox outbreak: A rapid review on comparative benefits and risks. Health Sci Rep 2022; 5:e798. [PMID: 36032515 PMCID: PMC9399446 DOI: 10.1002/hsr2.798] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/24/2022] [Accepted: 08/01/2022] [Indexed: 01/14/2023] Open
Abstract
Background and aims There is a sought for vaccines and antiviral agents as countermeasures for the recent monkeypox outbreak. Here, we aimed to review and discuss the repurposing potentials of smallpox vaccines and drugs in monkeypox outbreaks based on their comparative benefits and risks. Therefore, we conducted this rapid review and discussed the repurposing potentials of smallpox vaccines and drugs in monkeypox infection. Methods Here, we searched Google Scholar and PubMed for relevant information and data. We found many articles that have suggested the use of smallpox vaccines and antiviral drugs in monkeypox outbreaks according to the study findings. We read the relevant articles to extract information. Results According to the available documents, we found two replication-competent and one replication-deficient vaccinia vaccines were effective against Orthopoxvirus. However, the healthcare authorities have authorized second-generation live vaccina virus vaccines against Orthopoxvirus in many countries. Smallpox vaccine is almost 85% effective in preventing monkeypox infection as monkeypox virus, variola virus, and vaccinia virus are similar. The United States and Canada have approved a replication-deficient third-generation smallpox vaccine for the prevention of monkeypox infection. However, the widely used second-generation smallpox vaccines contain a live virus and replicate it into the human cell. Therefore, there is a chance to cause virus-induced complications among the vaccinated subjects. In those circumstances, the available Orthopoxvirus inhibitors might be a good choice for treating monkeypox infections as they showed similar efficacy in monkeypox infection in different animal model clinical trials. Also, the combined use of antiviral drugs and vaccinia immune globulin can enhance significant effectiveness in immunocompromised subjects. Conclusion Repurposing of these smallpox vaccines and antiviral agents might be weapons to fight monkeypox infection. Also, we recommend further investigations of smallpox vaccines and Orthopoxvirus inhibitors in a human model study to explore their exact role in human monkeypox infections.
Collapse
Affiliation(s)
| | | | - Arpira Roy
- Department of BiotechnologySharda UniversityGreater NoidaIndia
| | | | - Md. Ashrafur Rahman
- Department of Pharmaceutical SciencesJerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC)AmarilloTexasUSA
| | | | | |
Collapse
|
8
|
Madar-Balakirski N, Rosner A, Melamed S, Politi B, Steiner M, Tamir H, Yahalom-Ronen Y, Bar-David E, Ben-Shmuel A, Sittner A, Glinert I, Weiss S, Bar-Haim E, Cohen H, Elia U, Achdout H, Erez N, Rotem S, Lazar S, Nyska A, Yitzhaki S, Beth-Din A, Levy H, Paran N, Israely T, Marcus H. Preliminary nonclinical safety and immunogenicity of an rVSV-ΔG-SARS-CoV-2-S vaccine in mice, hamsters, rabbits and pigs. Arch Toxicol 2022; 96:859-875. [PMID: 35032184 PMCID: PMC8760087 DOI: 10.1007/s00204-021-03214-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/21/2021] [Indexed: 12/18/2022]
Abstract
rVSV-ΔG-SARS-CoV-2-S is a clinical stage (Phase 2) replication competent recombinant vaccine against SARS-CoV-2. To evaluate the safety profile of the vaccine, a series of non-clinical safety, immunogenicity and efficacy studies were conducted in four animal species, using multiple doses (up to 108 Plaque Forming Units/animal) and dosing regimens. There were no treatment-related mortalities or any noticeable clinical signs in any of the studies. Compared to unvaccinated controls, hematology and biochemistry parameters were unremarkable and no adverse histopathological findings. There was no detectable viral shedding in urine, nor viral RNA detected in whole blood or serum samples seven days post vaccination. The rVSV-ΔG-SARS-CoV-2-S vaccination gave rise to neutralizing antibodies, cellular immune responses, and increased lymphocytic cellularity in the spleen germinal centers and regional lymph nodes. No evidence for neurovirulence was found in C57BL/6 immune competent mice or in highly sensitive type I interferon knock-out mice. Vaccine virus replication and distribution in K18-human Angiotensin-converting enzyme 2-transgenic mice showed a gradual clearance from the vaccination site with no vaccine virus recovered from the lungs. The nonclinical data suggest that the rVSV-ΔG-SARS-CoV-2-S vaccine is safe and immunogenic. These results supported the initiation of clinical trials, currently in Phase 2.
Collapse
Affiliation(s)
- Noa Madar-Balakirski
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Amir Rosner
- Veterinary Center for Preclinical Research, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Sharon Melamed
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Boaz Politi
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | | | - Hadas Tamir
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Yfat Yahalom-Ronen
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Elad Bar-David
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Amir Ben-Shmuel
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Assa Sittner
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Itai Glinert
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shay Weiss
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Erez Bar-Haim
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Hila Cohen
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Uri Elia
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Hagit Achdout
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Noam Erez
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shahar Rotem
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shlomi Lazar
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Abraham Nyska
- Sackler School of Medicine, Tel Aviv University, and Consultant in Toxicologic Pathology, Tel Aviv, Israel
| | - Shmuel Yitzhaki
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Adi Beth-Din
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Haim Levy
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Nir Paran
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Tomer Israely
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel.
| | - Hadar Marcus
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona, Israel.
| |
Collapse
|
9
|
Modeling SARS-CoV-2 Infection in Mice Using Lentiviral hACE2 Vectors Infers Two Modes of Immune Responses to SARS-CoV-2 Infection. Viruses 2021; 14:v14010011. [PMID: 35062215 PMCID: PMC8778683 DOI: 10.3390/v14010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 11/18/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused a severe global pandemic. Mice models are essential to investigate infection pathology, antiviral drugs, and vaccine development. However, wild-type mice lack the human angiotensin-converting enzyme 2 (hACE2) that mediates SARS-CoV-2 entry into human cells and consequently are not susceptible to SARS-CoV-2 infection. hACE2 transgenic mice could provide an efficient COVID-19 model, but are not always readily available, and practically restricted to specific strains. Therefore, there is a dearth of additional mouse models for SARS-CoV-2 infection. We applied lentiviral vectors to generate hACE2 expression in interferon receptor knock-out (IFNAR1−/−) mice. Lenti-hACE2 transduction supported SARS-CoV-2 replication in vivo, simulating mild acute lung disease. Gene expression analysis revealed two modes of immune responses to SARS-CoV-2 infection: one in response to the exposure of mouse lungs to SARS-CoV-2 particles in the absence of productive viral replication, and the second in response to productive SARS-CoV-2 infection. Our results infer that immune response to immunogenic elements on incoming virus or in productively infected cells stimulate diverse immune effectors, even in absence of type I IFN signaling. Our findings should contribute to a better understanding of the immune response triggered by SARS-CoV-2 and to further elucidate COVID-19.
Collapse
|
10
|
Alkaloid-like annulated pyrano[4,3-b]pyrroles: antiviral activity and hydrolysis. Chem Heterocycl Compd (N Y) 2021. [DOI: 10.1007/s10593-021-02928-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
11
|
Pharmacokinetics and Efficacy of a Potential Smallpox Therapeutic, Brincidofovir, in a Lethal Monkeypox Virus Animal Model. mSphere 2021; 6:6/1/e00927-20. [PMID: 33536322 PMCID: PMC7860987 DOI: 10.1128/msphere.00927-20] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Smallpox, caused by Variola virus (VARV), was eradicated in 1980; however, VARV bioterrorist threats still exist, necessitating readily available therapeutics. Current preparedness activities recognize the importance of oral antivirals and recommend therapeutics with different mechanisms of action. Monkeypox virus (MPXV) is closely related to VARV, causing a highly similar clinical human disease, and can be used as a surrogate for smallpox antiviral testing. The prairie dog MPXV model has been characterized and used to study the efficacy of antipoxvirus therapeutics, including recently approved TPOXX (tecovirimat). Brincidofovir (BCV; CMX001) has shown antiviral activity against double-stranded DNA viruses, including poxviruses. To determine the exposure of BCV following oral administration to prairie dogs, a pharmacokinetics (PK) study was performed. Analysis of BCV plasma concentrations indicated variability, conceivably due to the outbred nature of the animals. To determine BCV efficacy in the MPXV prairie dog model, groups of animals were intranasally challenged with 9 × 105 plaque-forming units (PFU; 90% lethal dose [LD90]) of MPXV on inoculation day 0 (ID0). Animals were divided into groups based on the first day of BCV treatment relative to inoculation day (ID-1, ID0, or ID1). A trend in efficacy was noted dependent upon treatment initiation (57% on ID-1, 43% on ID0, and 29% on ID1) but was lower than demonstrated in other animal models. Analysis of the PK data indicated that BCV plasma exposure (maximum concentration [C max]) and the time of the last quantifiable concentration (AUClast) were lower than in other animal models administered the same doses, indicating that suboptimal BCV exposure may explain the lower protective effect on survival.IMPORTANCE Preparedness activities against highly transmissible viruses with high mortality rates have been highlighted during the ongoing coronavirus disease 2019 (COVID-19) pandemic. Smallpox, caused by variola virus (VARV) infection, is highly transmissible, with an estimated 30% mortality. Through an intensive vaccination campaign, smallpox was declared eradicated in 1980, and routine smallpox vaccination of individuals ceased. Today's current population has little/no immunity against VARV. If smallpox were to reemerge, the worldwide results would be devastating. Recent FDA approval of one smallpox antiviral (tecovirimat) was a successful step in biothreat preparedness; however, orthopoxviruses can become resistant to treatment, suggesting the need for multiple therapeutics. Our paper details the efficacy of the investigational smallpox drug brincidofovir in a monkeypox virus (MPXV) animal model. Since brincidofovir has not been tested in vivo against smallpox, studies with the related virus MPXV are critical in understanding whether it would be protective in the event of a smallpox outbreak.
Collapse
|
12
|
Identification and genetic characterization of a novel Orthobunyavirus species by a straightforward high-throughput sequencing-based approach. Sci Rep 2019; 9:3398. [PMID: 30833612 PMCID: PMC6399452 DOI: 10.1038/s41598-019-40036-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/07/2019] [Indexed: 12/04/2022] Open
Abstract
Identification and characterization of novel unknown viruses is of great importance. The introduction of high-throughput sequencing (HTS)-based methods has paved the way for genomics-based detection of pathogens without any prior assumptions about the characteristics of the organisms. However, the use of HTS for the characterization of viral pathogens from clinical samples remains limited. Here, we report the identification of a novel Orthobunyavirus species isolated from horse plasma. The identification was based on a straightforward HTS approach. Following enrichment in cell culture, RNA was extracted from the growth medium and rapid library preparation, HTS and primary bioinformatic analyses were performed in less than 12 hours. Taxonomical profiling of the sequencing reads did not reveal sequence similarities to any known virus. Subsequent application of de novo assembly tools to the sequencing reads produced contigs, of which three showed some similarity to the L, M, and S segments of viruses belonging to the Orthobunyavirus genus. Further refinement of these contigs resulted in high-quality, full-length genomic sequences of the three genomic segments (L, M and S) of a novel Orthobunyavirus. Characterization of the genomic sequence, including the prediction of open reading frames and the inspection of consensus genomic termini and phylogenetic analysis, further confirmed that the novel virus is indeed a new species, which we named Ness Ziona virus.
Collapse
|
13
|
Differential Response Following Infection of Mouse CNS with Virulent and Attenuated Vaccinia Virus Strains. Vaccines (Basel) 2019; 7:vaccines7010019. [PMID: 30759813 PMCID: PMC6466266 DOI: 10.3390/vaccines7010019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/04/2019] [Accepted: 02/07/2019] [Indexed: 12/18/2022] Open
Abstract
Viral infections of the central nervous system (CNS) lead to a broad range of pathologies. CNS infections with Orthopox viruses have been mainly documented as an adverse reaction to smallpox vaccination with vaccinia virus. To date, there is insufficient data regarding the mechanisms underlying pathological viral replication or viral clearance. Therefore, informed risk assessment of vaccine adverse reactions or outcome prediction is limited. This work applied a model of viral infection of the CNS, comparing neurovirulent with attenuated strains. We followed various parameters along the disease and correlated viral load, morbidity, and mortality with tissue integrity, innate and adaptive immune response and functionality of the blood–brain barrier. Combining these data with whole brain RNA-seq analysis performed at different time points indicated that neurovirulence is associated with host immune silencing followed by induction of tissue damage-specific pathways. In contrast, brain infection with attenuated strains resulted in rapid and robust induction of innate and adaptive protective immunity, followed by viral clearance and recovery. This study significantly improves our understanding of the mechanisms and processes determining the consequence of viral CNS infection and highlights potential biomarkers associated with such outcomes.
Collapse
|
14
|
Green MS, LeDuc J, Cohen D, Franz DR. Confronting the threat of bioterrorism: realities, challenges, and defensive strategies. THE LANCET. INFECTIOUS DISEASES 2018; 19:e2-e13. [PMID: 30340981 PMCID: PMC7106434 DOI: 10.1016/s1473-3099(18)30298-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/25/2018] [Accepted: 05/04/2018] [Indexed: 01/30/2023]
Abstract
Global terrorism is a rapidly growing threat to world security, and increases the risk of bioterrorism. In this Review, we discuss the potential threat of bioterrorism, agents that could be exploited, and recent developments in technologies and policy for detecting and controlling epidemics that have been initiated intentionally. The local and international response to infectious disease epidemics, such as the severe acute respiratory syndrome and west African Ebola virus epidemic, revealed serious shortcomings which bioterrorists might exploit when intentionally initiating an epidemic. Development of new vaccines and antimicrobial therapies remains a priority, including the need to expedite clinical trials using new methodologies. Better means to protect health-care workers operating in dangerous environments are also needed, particularly in areas with poor infrastructure. New and improved approaches should be developed for surveillance, early detection, response, effective isolation of patients, control of the movement of potentially infected people, and risk communication. Access to dangerous pathogens should be appropriately regulated, without reducing progress in the development of countermeasures. We conclude that preparedness for intentional outbreaks has the important added value of strengthening preparedness for natural epidemics, and vice versa.
Collapse
Affiliation(s)
- Manfred S Green
- School of Public Health, University of Haifa, Haifa, Israel.
| | - James LeDuc
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
| | - Daniel Cohen
- School of Public Health, Tel Aviv University, Tel Aviv, Israel
| | - David R Franz
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
15
|
Melamed S, Israely T, Paran N. Challenges and Achievements in Prevention and Treatment of Smallpox. Vaccines (Basel) 2018; 6:vaccines6010008. [PMID: 29382130 PMCID: PMC5874649 DOI: 10.3390/vaccines6010008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/15/2018] [Accepted: 01/26/2018] [Indexed: 01/17/2023] Open
Abstract
Declaration of smallpox eradication by the WHO in 1980 led to discontinuation of the worldwide vaccination campaign. The increasing percentage of unvaccinated individuals, the existence of its causative infectious agent variola virus (VARV), and the recent synthetic achievements increase the threat of intentional or accidental release and reemergence of smallpox. Control of smallpox would require an emergency vaccination campaign, as no other protective measure has been approved to achieve eradication and ensure worldwide protection. Experimental data in surrogate animal models support the assumption, based on anecdotal, uncontrolled historical data, that vaccination up to 4 days postexposure confers effective protection. The long incubation period, and the uncertainty of the exposure status in the surrounding population, call for the development and evaluation of safe and effective methods enabling extension of the therapeutic window, and to reduce the disease manifestations and vaccine adverse reactions. To achieve these goals, we need to evaluate the efficacy of novel and already licensed vaccines as a sole treatment, or in conjunction with immune modulators and antiviral drugs. In this review, we address the available data, recent achievements, and open questions.
Collapse
Affiliation(s)
- Sharon Melamed
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness-Ziona 74100, Israel.
| | - Tomer Israely
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness-Ziona 74100, Israel.
| | - Nir Paran
- Department of Infectious Diseases, Israel Institute for Biological Research, P.O. Box 19, Ness-Ziona 74100, Israel.
| |
Collapse
|
16
|
Israely T, Melamed S, Achdout H, Erez N, Politi B, Waner T, Lustig S, Paran N. TLR3 and TLR9 agonists improve postexposure vaccination efficacy of live smallpox vaccines. PLoS One 2014; 9:e110545. [PMID: 25350003 PMCID: PMC4211728 DOI: 10.1371/journal.pone.0110545] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 09/14/2014] [Indexed: 12/14/2022] Open
Abstract
Eradication of smallpox and discontinuation of the vaccination campaign resulted in an increase in the percentage of unvaccinated individuals, highlighting the need for postexposure efficient countermeasures in case of accidental or deliberate viral release. Intranasal infection of mice with ectromelia virus (ECTV), a model for human smallpox, is curable by vaccination with a high vaccine dose given up to 3 days postexposure. To further extend this protective window and to reduce morbidity, mice were vaccinated postexposure with Vaccinia-Lister, the conventional smallpox vaccine or Modified Vaccinia Ankara, a highly attenuated vaccine in conjunction with TLR3 or TLR9 agonists. We show that co-administration of the TLR3 agonist poly(I:C) even 5 days postexposure conferred protection, avoiding the need to increase the vaccination dose. Efficacious treatments prevented death, ameliorated disease symptoms, reduced viral load and maintained tissue integrity of target organs. Protection was associated with significant elevation of serum IFNα and anti-vaccinia IgM antibodies, modulation of IFNγ response, and balanced activation of NK and T cells. TLR9 agonists (CpG ODNs) were less protective than the TLR3 agonist poly(I:C). We show that activation of type 1 IFN by poly(I:C) and protection is achievable even without co-vaccination, requiring sufficient amount of the viral antigens of the infective agent or the vaccine. This study demonstrated the therapeutic potential of postexposure immune modulation by TLR activation, allowing to alleviate the disease symptoms and to further extend the protective window of postexposure vaccination.
Collapse
Affiliation(s)
- Tomer Israely
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Sharon Melamed
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Hagit Achdout
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Noam Erez
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Boaz Politi
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Trevor Waner
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Shlomo Lustig
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Nir Paran
- Department of Infectious diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
- * E-mail:
| |
Collapse
|
17
|
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: 1.7] [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
|
18
|
Current concepts in the management of biologic and chemical warfare causalities. J Trauma Acute Care Surg 2013; 75:582-9. [PMID: 24064869 DOI: 10.1097/ta.0b013e3182a11175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
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: 1.5] [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
|