1
|
Butic AB, Katz ZE, Jin G, Fukushima K, Hazama M, Lukacher AE, Lauver MD. Brincidofovir inhibits polyomavirus infection in vivo. mBio 2024; 15:e0104924. [PMID: 38953354 PMCID: PMC11323531 DOI: 10.1128/mbio.01049-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/06/2024] [Indexed: 07/04/2024] Open
Abstract
Polyomaviruses are species-specific DNA viruses that can cause disease in immunocompromised individuals. Despite their role as the causative agents for several diseases, there are no currently approved antivirals for treating polyomavirus infection. Brincidofovir (BCV) is an antiviral approved for the treatment of poxvirus infections and has shown activity against other double-stranded DNA viruses. In this study, we tested the efficacy of BCV against polyomavirus infection in vitro and in vivo using mouse polyomavirus (MuPyV). BCV inhibited virus production in primary mouse kidney cells and brain cortical cells. BCV treatment of cells transfected with MuPyV genomic DNA resulted in a reduction in virus levels, indicating that viral inhibition occurs post-entry. Although in vitro BCV treatment had a limited effect on viral DNA and RNA levels, drug treatment was associated with a reduction in viral protein, raising the possibility that BCV acts post-transcriptionally to inhibit MuPyV infection. In mice, BCV treatment was well tolerated, and prophylactic treatment resulted in a reduction in viral DNA levels and a potent suppression of infectious virus production in the kidney and brain. In mice with chronic polyomavirus infection, therapeutic administration of BCV decreased viremia and reduced infection in the kidney. These data demonstrate that BCV exerts antiviral activity against polyomavirus infection in vivo, supporting further investigation into the use of BCV to treat clinical polyomavirus infections. IMPORTANCE Widespread in the human population and able to persist asymptomatically for the life of an individual, polyomavirus infections cause a significant disease burden in the immunocompromised. Individuals undergoing immune suppression, such as kidney transplant patients or those treated for autoimmune diseases, are particularly at high risk for polyomavirus-associated diseases. Because no antiviral agent exists for treating polyomavirus infections, management of polyomavirus-associated diseases typically involves reducing or discontinuing immunomodulatory therapy. This can be perilous due to the risk of transplant rejection and the potential development of adverse immune reactions. Thus, there is a pressing need for the development of antivirals targeting polyomaviruses. Here, we investigate the effects of brincidofovir, an FDA-approved antiviral, on polyomavirus infection in vivo using mouse polyomavirus. We show that the drug is well-tolerated in mice, reduces infectious viral titers, and limits viral pathology, indicating the potential of brincidofovir as an anti-polyomavirus therapeutic.
Collapse
Affiliation(s)
- Arrienne B. Butic
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Zoe E. Katz
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Ge Jin
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Koji Fukushima
- SymBio Pharmaceuticals Limited, Toranomon, Minato, Tokyo, Japan
| | | | - Aron E. Lukacher
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Matthew D. Lauver
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
2
|
Tollefson AE, Cline-Smith A, Spencer JF, Ying B, Reyna DM, Lipka E, James SH, Toth K. Longitudinal Monitoring of the Effects of Anti-Adenoviral Treatment Regimens in a Permissive In Vivo Model. Viruses 2024; 16:1200. [PMID: 39205174 PMCID: PMC11359180 DOI: 10.3390/v16081200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Adenovirus infections of immunocompromised patients can cause life-threatening disseminated disease. While there are presently no drugs specifically approved to treat these infections, there are several compounds that showed efficacy against adenovirus in preclinical studies. For any such compound, low toxicity is an essential requirement. As cumulative drug effects can accentuate pathology, especially in patients with other morbidities, it is important to limit antiviral exposure to what is absolutely necessary. This is achievable by monitoring the virus burden of the patients and administering antivirals to suppress virus replication to a non-pathogenic level. We modeled such a system using Syrian hamsters infected with a replication-competent adenovirus vector, in which luciferase expression is coupled to virus replication. We found that virus replication could be followed in vivo in the same animal by repeated measurement of luciferase expression. To test the utility of an interrupted treatment regimen, we used NPP-669 and valganciclovir, two antiviral compounds with high and moderate anti-adenoviral efficacy, respectively. We found that short-term treatment of adenovirus-infected hamsters at times of peak virus replication can prevent virus-associated pathology. Thus, we believe that this animal model can be used to model different treatment regimens for anti-adenoviral compounds.
Collapse
Affiliation(s)
- Ann E Tollefson
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Anna Cline-Smith
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Jacqueline F Spencer
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Baoling Ying
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | | | | | - Scott H James
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Karoly Toth
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| |
Collapse
|
3
|
Tollefson AE, Cline-Smith AB, Spencer JF, Reyna DM, Lipka E, Toth K. NPP-669, a prodrug of cidofovir, is highly efficacious against human adenovirus infection in the permissive Syrian hamster model. Antimicrob Agents Chemother 2024; 68:e0048924. [PMID: 38775484 PMCID: PMC11232382 DOI: 10.1128/aac.00489-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 04/29/2024] [Indexed: 07/10/2024] Open
Abstract
Human adenoviruses can cause serious, disseminated infections in immunocompromised patients. For pediatric allogeneic stem cell transplant patients, the case fatality rate can reach 80%. Still, there is no available antiviral drug that is specifically approved by the Food and Drug Administration for the treatment of adenovirus infections. To fill this pressing medical need, we have developed NPP-669, a prodrug of cidofovir with broad activity against double-stranded DNA viruses, including adenoviruses. Here, we report on the in vivo anti-adenoviral efficacy of NPP-669. Using the immunosuppressed Syrian hamster as the model, we show that NPP-669 is highly efficacious when dosed orally at 1 mg/kg and 3 mg/kg. In a delayed administration experiment, NPP-669 was more effective than brincidofovir, a similar compound that reached Phase III clinical trials. Furthermore, parenteral administration of NPP-669 increased its efficacy approximately 10-fold compared to oral dosing without apparent toxicity, suggesting that this route may be preferable in a hospital setting. Based on these findings, we believe that NPP-669 is a promising new compound that needs to be further investigated.
Collapse
Affiliation(s)
- Ann E Tollefson
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Anna B Cline-Smith
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Jacqueline F Spencer
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | | | | | - Karoly Toth
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
4
|
Palazzotti D, Sguilla M, Manfroni G, Cecchetti V, Astolfi A, Barreca ML. Small Molecule Drugs Targeting Viral Polymerases. Pharmaceuticals (Basel) 2024; 17:661. [PMID: 38794231 PMCID: PMC11124969 DOI: 10.3390/ph17050661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Small molecules that specifically target viral polymerases-crucial enzymes governing viral genome transcription and replication-play a pivotal role in combating viral infections. Presently, approved polymerase inhibitors cover nine human viruses, spanning both DNA and RNA viruses. This review provides a comprehensive analysis of these licensed drugs, encompassing nucleoside/nucleotide inhibitors (NIs), non-nucleoside inhibitors (NNIs), and mutagenic agents. For each compound, we describe the specific targeted virus and related polymerase enzyme, the mechanism of action, and the relevant bioactivity data. This wealth of information serves as a valuable resource for researchers actively engaged in antiviral drug discovery efforts, offering a complete overview of established strategies as well as insights for shaping the development of next-generation antiviral therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Maria Letizia Barreca
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06123 Perugia, Italy; (D.P.); (M.S.); (G.M.); (V.C.); (A.A.)
| |
Collapse
|
5
|
Wang J, Yang C, Liang Z, Sun J, Zhang M, Qiu S, Du X, He X, Pang X, Ma X, Xie M, Han X, Fan R, Zhou E, Yu H, She D, Song H, Wang J. Indirubin-3'-monoxime exhibits potent antiviral and anti-inflammatory effects against human adenoviruses in vitro and in vivo. Biomed Pharmacother 2024; 174:116558. [PMID: 38603887 DOI: 10.1016/j.biopha.2024.116558] [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: 12/25/2023] [Revised: 03/28/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024] Open
Abstract
Human adenovirus (HAdV) infection is a major cause of respiratory disease, yet no antiviral drugs have been approved for its treatment. Herein, we evaluated the antiviral and anti-inflammatory effects of cyclin-dependent protein kinase (CDK) inhibitor indirubin-3'-monoxime (IM) against HAdV infection in cells and a transgenic mouse model. After evaluating its cytotoxicity, cytopathic effect reduction, antiviral replication kinetics, and viral yield reduction assays were performed to assess the anti-HAdV activity of IM. Quantitative real-time polymerase chain reaction (qPCR), quantitative reverse transcription PCR (qRT-PCR), and western blotting were used to assess the effects of IM on HAdV DNA replication, transcription, and protein expression, respectively. IM significantly inhibited HAdV DNA replication as well as E1A and Hexon transcription, in addition to significantly suppressing the phosphorylation of the RNA polymerase II C-terminal domain (CTD). IM mitigated body weight loss, reduced viral burden, and lung injury, decreasing cytokine and chemokine secretion to a greater extent than cidofovir. Altogether, IM inhibits HAdV replication by downregulating CTD phosphorylation to suppress viral infection and corresponding innate immune reactions as a promising therapeutic agent.
Collapse
Affiliation(s)
- Junyu Wang
- Medical School of Chinese PLA, Beijing 100853, China; Department of Respiratory and Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Chaojie Yang
- Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| | - Zhixin Liang
- Department of Respiratory and Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Junping Sun
- Department of Respiratory and Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Mingyue Zhang
- Department of Respiratory and Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Shaofu Qiu
- Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| | - Xinying Du
- Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| | - Xi He
- Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| | - Xiaoying Pang
- Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| | - Xidong Ma
- Department of Respiratory and Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Mei Xie
- Department of Respiratory and Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Xinjie Han
- Department of Respiratory and Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Ru Fan
- Department of Respiratory and Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Enlu Zhou
- Department of Respiratory and Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Hairong Yu
- Department of Respiratory Medicine, 71st Group Military Hospital of PLA Army, Xuzhou, Jiangsu Province 221004, China
| | - Danyang She
- Department of Respiratory and Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China.
| | - Hongbin Song
- Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China.
| | - Jianxin Wang
- Department of Respiratory and Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
6
|
Gupta A, Khanna P, Parihar A, Singh DP, Singhi SC. Cidofovir in Severe Hypoxemic Adenoviral Pneumonia. Indian J Pediatr 2024; 91:398-400. [PMID: 37620591 DOI: 10.1007/s12098-023-04812-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023]
Abstract
The authors present a 16-mo-old boy with flu like symptoms, not responding to supportive management and progressed to severe hypoxemic pneumonia. Adenovirus was detected in the nasopharyngeal aspirate. He showed rapid improvement after intravenous cidofovir administration.
Collapse
Affiliation(s)
- Aditi Gupta
- Department of Pediatrics, Amrita Hospital, Mata Amritanandamayi Marg, Sector 88, Faridabad, Haryana, 121002, India
| | - Pooja Khanna
- Department of Pediatrics, Amrita Hospital, Mata Amritanandamayi Marg, Sector 88, Faridabad, Haryana, 121002, India
| | - Arjun Parihar
- Department of Pediatrics, Amrita Hospital, Mata Amritanandamayi Marg, Sector 88, Faridabad, Haryana, 121002, India
| | - Dhirendra Pratap Singh
- Department of Pediatric Critical Care, Amrita Hospital, Mata Amritanandamayi Marg, Sector 88, Faridabad, Haryana, 121002, India
| | - Sunit Chandra Singhi
- Department of Pediatrics, Amrita Hospital, Mata Amritanandamayi Marg, Sector 88, Faridabad, Haryana, 121002, India.
| |
Collapse
|
7
|
Becken BA, Lamson DM, Gonzalez G, Patel S, St. George K, Kajon AE. A Fulminant Case of Adenovirus Genotype C108 Infection in a Pediatric Stem Cell Transplant Recipient with x-Linked Lymphoproliferative Syndrome Type 1. Viruses 2024; 16:137. [PMID: 38257837 PMCID: PMC10819400 DOI: 10.3390/v16010137] [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: 01/02/2024] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
A 3-year-old male with X-linked lymphoproliferative syndrome type 1 underwent an unrelated umbilical cord blood transplant (UUCBT). The week prior to transplant the patient tested positive for adenovirus (HAdV) with a viral load of <190 copies/mL and was started on cidofovir. UUCBT proceeded as scheduled, and the patient engrafted on day +19. The patient's HAdV load in serum continued to rise with resulting hepatic dysfunction, despite ongoing therapy with cidofovir and HAdV specific T-cell infusions. The patient died 6 months after transplantation having never cleared the virus. Next generation whole genome sequencing and sequence data analyses identified an intertypic recombinant HAdV-C P1H2F2 closely related (99.6% similarity) to genotype C108 in the isolates from three blood specimens obtained during the last week of life. Incidentally, the de novo assembly strategy enabled the detection of an adeno-associated virus type 2 (AAV2) genome in the DNA purified from the plasma isolates. Proteotyping analysis revealed minor differences in the predicted amino acid sequences for E1A, E1B 19K, E1B 55K, DNA polymerase, penton base, and fiber. None of the mutations previously described for HAdV-C5 variants resistant to cidofovir were identified. In silico restriction enzyme analysis revealed a distinct Sac I profile for the identified virus, supporting its designation as a C108 variant.
Collapse
Affiliation(s)
- Bradford A. Becken
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA; (B.A.B.); (S.P.)
| | - Daryl M. Lamson
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA; (D.M.L.); (K.S.G.)
| | - Gabriel Gonzalez
- UCD National Virus Reference Laboratory, Dublin, Ireland;
- Japan Initiative for World-Leading Vaccine Research and Development Centers, Institute for Vaccine Research and Development, Hokkaido University, Hokkaido, Japan
| | - Sachit Patel
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA; (B.A.B.); (S.P.)
| | - Kirsten St. George
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA; (D.M.L.); (K.S.G.)
| | - Adriana E. Kajon
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA
| |
Collapse
|
8
|
Shamim MA, Satapathy P, Padhi BK, Veeramachaneni SD, Akhtar N, Pradhan A, Agrawal A, Dwivedi P, Mohanty A, Pradhan KB, Kabir R, Rabaan AA, Alotaibi J, Al Ismail ZA, Alsoliabi ZA, Al Fraij A, Sah R, Rodriguez-Morales AJ. Pharmacological treatment and vaccines in monkeypox virus: a narrative review and bibliometric analysis. Front Pharmacol 2023; 14:1149909. [PMID: 37214444 PMCID: PMC10196034 DOI: 10.3389/fphar.2023.1149909] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Mpox (earlier known as monkeypox) virus infection is a recognized public health emergency. There has been little research on the treatment options. This article reviews the specific drugs used to treat mpox virus infection and the vaccines used here. Instead of focusing on the mechanistic basis, this review narrates the practical, real-life experiences of individual patients of mpox virus disease being administered these medicines. We conducted a bibliometric analysis on the treatment of the mpox virus using data from several databases like PubMed, Scopus, and Embase. The research on this topic has grown tremendously recently but it is highly concentrated in a few countries. Cidofovir is the most studied drug. This is because it is indicated and also used off-label for several conditions. The drugs used for mpox virus infection include tecovirimat, cidofovir, brincidofovir, vaccinia immune globulin, and trifluridine. Tecovirimat is used most frequently. It is a promising option in progressive mpox disease in terms of both efficacy and safety. Brincidofovir has been associated with treatment discontinuation due to elevated hepatic enzymes. Cidofovir is also not the preferred drug, often used because of the unavailability of tecovirimat. Trifluridine is used topically as an add-on agent along with tecovirimat for ocular manifestations of mpox virus disease. No study reports individual patient data for vaccinia immune globulin. Though no vaccine is currently approved for mpox virus infection, ACAM 2000 and JYNNEOS are the vaccines being mainly considered. ACAM 2000 is capable of replicating and may cause severe adverse reactions. It is used when JYNNEOS is contraindicated. Several drugs and vaccines are under development and have been discussed alongside pragmatic aspects of mpox virus treatment and prevention. Further studies can provide more insight into the safety and efficacy of Tecovirimat in actively progressing mpox virus disease.
Collapse
Affiliation(s)
| | - Prakisini Satapathy
- Department of Virology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bijaya Kumar Padhi
- Department of Community Medicine, School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Naushaba Akhtar
- Indian Council of Medical Research—Regional Medical Research Centre, Bhubaneswar, India
| | - Anindita Pradhan
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, India
| | - Abhimanyu Agrawal
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, India
| | - Pradeep Dwivedi
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, India
- Centre of Excellence for Tribal Health, All India Institute of Medical Sciences, Jodhpur, India
| | - Aroop Mohanty
- All India Institute of Medical Sciences, Gorakhpur, India
| | | | - Russell Kabir
- School of Allied Health, Anglia Ruskin University, Essex, United Kingdom
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
| | - Jawaher Alotaibi
- Infectious Diseases Unit, King Faisal Specialist Hospital and Research Center, Department of Medicine, Riyadh, Saudi Arabia
| | - Zainab A. Al Ismail
- Long Term Care Department, Dhahran Long Term Hospital, Dhahran, Saudi Arabia
| | | | - Ali Al Fraij
- Medical Laboratories and Blood Bank Department, Jubail Health Network, Jubail, Saudi Arabia
| | - Ranjit Sah
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal
- Harvard Medical School, Boston, MA, United States
- Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Alfonso J. Rodriguez-Morales
- Faculty of Health Sciences, Universidad Científica del Sur, Lima, Peru
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| |
Collapse
|
9
|
Jennings MR, Parks RJ. Human Adenovirus Gene Expression and Replication Is Regulated through Dynamic Changes in Nucleoprotein Structure throughout Infection. Viruses 2023; 15:161. [PMID: 36680201 PMCID: PMC9863843 DOI: 10.3390/v15010161] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
Human adenovirus (HAdV) is extremely common and can rapidly spread in confined populations such as daycare centers, hospitals, and retirement homes. Although HAdV usually causes only minor illness in otherwise healthy patients, HAdV can cause significant morbidity and mortality in certain populations, such as the very young, very old, or immunocompromised individuals. During infection, the viral DNA undergoes dramatic changes in nucleoprotein structure that promote the rapid expression of viral genes, replication of the DNA, and generation of thousands of new infectious virions-each process requiring a distinct complement of virus and host-encoded proteins. In this review, we summarize our current understanding of the nucleoprotein structure of HAdV DNA during the various phases of infection, the cellular proteins implicated in mediating these changes, and the role of epigenetics in HAdV gene expression and replication.
Collapse
Affiliation(s)
- Morgan R. Jennings
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Robin J. Parks
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Department of Medicine, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
10
|
Rabaan AA, Abas AH, Tallei TE, Al-Zaher MA, Al-Sheef NM, Fatimawali, Al-Nass EZ, Al-Ebrahim EA, Effendi Y, Idroes R, Alhabib MF, Al-Fheid HA, Adam AA, Bin Emran T. Monkeypox outbreak 2022: What we know so far and its potential drug targets and management strategies. J Med Virol 2023; 95:e28306. [PMID: 36372558 DOI: 10.1002/jmv.28306] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/28/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2022]
Abstract
Monkeypox is a rare zoonotic disease caused by infection with the monkeypox virus. The disease can result in flu-like symptoms, fever, and a persistent rash. The disease is currently spreading throughout the world and prevention and treatment efforts are being intensified. Although there is no treatment that has been specifically approved for monkeypox virus infection, infected patients may benefit from using certain antiviral medications that are typically prescribed for the treatment of smallpox. The drugs are tecovirimat, brincidofovir, and cidofovir, all of which are currently in short supply due to the spread of the monkeypox virus. Resistance is also a concern, as widespread replication of the monkeypox virus can lead to mutations that produce monkeypox viruses that are resistant to the currently available treatments. This article discusses monkeypox disease, potential drug targets, and management strategies to overcome monkeypox disease. With the discovery of new drugs, it is hoped that the problem of insufficient drugs will be resolved, and it is not anticipated that drug resistance will become a major issue in the near future.
Collapse
Affiliation(s)
- Ali A Rabaan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.,Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia.,Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| | - Abdul Hawil Abas
- Faculty of Bioscience and Engineering, Ghent University, Ghent, Belgium
| | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado, North Sulawesi, Indonesia
| | - Mona A Al-Zaher
- Department of Commitment management, Directorate of Health Affairs in the Eastern Province, Dammam, Saudi Arabia
| | - Noor M Al-Sheef
- Department of Commitment management, Directorate of Health Affairs in the Eastern Province, Dammam, Saudi Arabia
| | - Fatimawali
- Pharmacy Study Program, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado, North Sulawesi, Indonesia
| | - Esraa Z Al-Nass
- Department of Commitment management, Directorate of Health Affairs in the Eastern Province, Dammam, Saudi Arabia
| | - Eba A Al-Ebrahim
- Department of Commitment management, Directorate of Health Affairs in the Eastern Province, Dammam, Saudi Arabia
| | - Yunus Effendi
- Department of Biology, Faculty of Science and Technology, Al-Azhar Indonesia University, Jakarta, Indonesia
| | - Rinaldi Idroes
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Mather F Alhabib
- Molecular Diagnostic Laboratory, Dammam Regional Laboratory and Blood Bank, Dammam, Saudi Arabia
| | - Hussain A Al-Fheid
- Molecular Diagnostic Laboratory, Dammam Regional Laboratory and Blood Bank, Dammam, Saudi Arabia
| | - Ahmad Akroman Adam
- Dentistry Study Program, Faculty of Medicine, Sam Ratulangi University, Manado, North Sulawesi, Indonesia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh.,Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| |
Collapse
|
11
|
Li H, Huang QZ, Zhang H, Liu ZX, Chen XH, Ye LL, Luo Y. The land-scape of immune response to monkeypox virus. EBioMedicine 2022; 87:104424. [PMID: 36584594 PMCID: PMC9797195 DOI: 10.1016/j.ebiom.2022.104424] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/11/2022] [Accepted: 12/06/2022] [Indexed: 12/29/2022] Open
Abstract
Human monkeypox is a viral zoonotic smallpox-like disease caused by the monkeypox virus (MPXV) and has become the greatest public health threat in the genus Orthopoxvirus after smallpox was eradicated. The host immune response to MPXV plays an essential role in disease pathogenesis and clinical manifestations. MPXV infection leads to skin lesions with the genital area as the main feature in the current outbreak and triggers a strong immune response that results in sepsis, deep tissue abscess, severe respiratory disease, and injuries to multiple immune organs. Emerging evidence shows that the immunopathogenesis of MPXV infection is closely associated with impaired NK-cell function, lymphopenia, immune evasion, increased antibodies, increased blood monocytes and granulocytes, cytokine storm, inhibition of the host complement system, and antibody-dependent enhancement. In this overview, we discuss the immunopathology and immunopathogenesis of monkeypox to aid the development of novel immunotherapeutic strategies against monkeypox.
Collapse
Affiliation(s)
- Heng Li
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400044, PR China
| | - Qi-Zhao Huang
- Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Hong Zhang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 250033, Jinan, Shandong, China
| | - Zhen-Xing Liu
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400044, PR China
| | - Xiao-Hui Chen
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400044, PR China
| | - Li-Lin Ye
- Institute of Immunology, Third Military Medical University, Chongqing, 400038, PR China,Corresponding author: Institute of Immunology, Third Military Medical University, Chongqing, 400038, PR China.
| | - Yang Luo
- College of Life Sciences and Laboratory Medicine, Kunming Medical University, Kunming, Yunnan, 650500, PR China,Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, 650118, PR China,Department of Laboratory Medicine, Jiangjin Hospital, Chongqing University, Chongqing, 402260, PR China,Corresponding author: College of Life Sciences and Laboratory Medicine, Kunming Medical University, Kunming, Yunnan, 650500, PR China.
| |
Collapse
|
12
|
Stevaert A, Groaz E, Naesens L. Nucleoside analogs for management of respiratory virus infections: mechanism of action and clinical efficacy. Curr Opin Virol 2022; 57:101279. [PMID: 36403338 PMCID: PMC9671222 DOI: 10.1016/j.coviro.2022.101279] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 11/18/2022]
Abstract
The COVID-19 pandemic has accelerated the development of nucleoside analogs to treat respiratory virus infections, with remdesivir being the first compound to receive worldwide authorization and three other nucleoside analogs (i.e. favipiravir, molnupiravir, and bemnifosbuvir) in the pipeline. Here, we summarize the current knowledge concerning their clinical efficacy in suppressing the virus and reducing the need for hospitalization or respiratory support. We also mention trials of favipiravir and lumicitabine, for influenza and respiratory syncytial virus, respectively. Besides, we outline how nucleoside analogs interact with the polymerases of respiratory viruses, to cause lethal virus mutagenesis or disturbance of viral RNA synthesis. In this way, we aim to convey the key findings on this rapidly evolving class of respiratory virus medication.
Collapse
Affiliation(s)
- Annelies Stevaert
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Herestraat 49 box 1043, B-3000 Leuven, Belgium
| | - Elisabetta Groaz
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49 box 1041, B-3000 Leuven, Belgium; Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Lieve Naesens
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Herestraat 49 box 1043, B-3000 Leuven, Belgium.
| |
Collapse
|
13
|
Johri N, Kumar D, Nagar P, Maurya A, Vengat M, Jain P. Clinical manifestations of human monkeypox infection and implications for outbreak strategy. HEALTH SCIENCES REVIEW (OXFORD, ENGLAND) 2022; 5:100055. [PMID: 36254190 PMCID: PMC9535997 DOI: 10.1016/j.hsr.2022.100055] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/08/2022] [Accepted: 10/02/2022] [Indexed: 11/09/2022]
Abstract
Monkeypox is an orthopoxvirus-based zoonotic illness that causes symptoms similar to smallpox in humans. Health care workers around the world are making it a priority to educate themselves on the many clinical manifestations and treatment options for this virus as public health agencies strive to stop the current outbreak. The infected do not have access to any treatment at this time. However, information obtained from the smallpox pandemic has led researchers to examine vaccinia immune globulin (IVG), tecovirimat, and cidofovir as viable treatments for monkeypox. Moreover, medication like tecovirimat may be given in extreme circumstances, and supportive therapy can help with symptom relief. The European Medicines Agency (EMA) certified tecovirimat as safe and effective against monkeypox in 2022, per the World Health Organization (WHO). As there are now no established guidelines for alleviating these symptoms, the efficacy of these treatments is highly questionable. Some high-profile cases in recent years have cast doubt on the long-held belief that this illness is rare and always resolves itself without treatment. We aimed to conduct this review to get a deeper comprehension of the evolving epidemiology of monkeypox by analysing such factors as the number of confirmed, probable, and potential cases, the median age at presentation, the mortality rate, and the geographic distribution of the disease. This study offers an updated review of monkeypox and the clinical treatments that are currently available as a result of the worldwide epidemics.
Collapse
Affiliation(s)
- Nishant Johri
- Department of Pharmacy Practice, Teerthanker Mahaveer Hospital & Research Centre, Moradabad, Uttar Pradesh, India
| | - Deepanshu Kumar
- Department of Pharmacy Practice, Teerthanker Mahaveer Hospital & Research Centre, Moradabad, Uttar Pradesh, India
| | - Priya Nagar
- Department of Pharmacy Practice, Teerthanker Mahaveer Hospital & Research Centre, Moradabad, Uttar Pradesh, India
| | - Aditya Maurya
- Department of Pharmacy Practice, Teerthanker Mahaveer Hospital & Research Centre, Moradabad, Uttar Pradesh, India
| | - Maheshwari Vengat
- Department of Oncology, Chester Medical School, Chester, United Kingdom
| | - Parag Jain
- Department of Pharmacology, Chhatrapati Shivaji Institute of Pharmacy, Durg, Chhattisgarh 491001, India
| |
Collapse
|
14
|
The evolving epidemiology of monkeypox virus. Cytokine Growth Factor Rev 2022; 68:1-12. [PMID: 36244878 PMCID: PMC9547435 DOI: 10.1016/j.cytogfr.2022.10.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 02/07/2023]
Abstract
Monkeypox, caused by the monkeypox virus (MPXV), is a zoonotic disease endemic mainly in West and Central Africa. As of 27 September 2022, human monkeypox has occurred in more than 100 countries (mostly in non-endemic regions) and caused over 66,000 confirmed cases, which differs from previous epidemics that mainly affected African countries. Due to the increasing number of confirmed cases worldwide, the World Health Organization (WHO) has declared the monkeypox outbreak as a Public Health Emergency of International Concern on July 23, 2022. The international outbreak of human monkeypox represents a novel route of transmission for MPXV, with genital lesions as the primary infection, and the emergence of monkeypox in the current outbreak is also new, as novel variants emerge. Clinical physicians and scientists should be aware of this emerging situation, which presents a different scenario from previous outbreaks. In this review, we will discuss the molecular virology, evasion of antiviral immunity, epidemiology, evolution, and detection of MPXV, as well as prophylaxis and treatment strategies for monkeypox. This review also emphasizes the integration of relevant epidemiological data with genomic surveillance data to obtain real-time data, which could formulate prevention and control measures to curb this outbreak.
Collapse
|
15
|
Mazzotta S, Berastegui-Cabrera J, Carullo G, Vega-Holm M, Carretero-Ledesma M, Mendolia L, Aiello F, Iglesias-Guerra F, Pachón J, Vega-Pérez JM, Sánchez-Céspedes J. Serinol-Based Benzoic Acid Esters as New Scaffolds for the Development of Adenovirus Infection Inhibitors: Design, Synthesis, and In Vitro Biological Evaluation. ACS Infect Dis 2021; 7:1433-1444. [PMID: 33073569 DOI: 10.1021/acsinfecdis.0c00515] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the years, human adenovirus (HAdV) has progressively been recognized as a significant viral pathogen. Traditionally associated with self-limited respiratory, gastrointestinal, and conjunctival infections, mainly in immunocompromised patients, HAdV is currently considered to be a pathogen presenting significant morbidity and mortality in both immunosuppressed and otherwise healthy individuals. Currently available therapeutic options are limited because of their lack of effectivity and related side effects. In this context, there is an urgent need to develop effective anti-HAdV drugs with suitable therapeutic indexes. In this work, we identified new serinol-derived benzoic acid esters as novel scaffolds for the inhibition of HAdV infections. A set of 38 compounds were designed and synthesized, and their antiviral activity and cytotoxicity were evaluated. Four compounds (13, 14, 27, and 32) inhibited HAdV infection at low micromolar concentrations (2.82-5.35 μM). Their half maximal inhibitory concentration (IC50) values were lower compared to that of cidofovir, the current drug of choice. All compounds significantly reduced the HAdV DNA replication process, while they did not block any step of the viral entry. Our results showed that compounds 13, 14, and 32 seem to be targeting the expression of the E1A early gene. Moreover, all four derivatives demonstrated a significant inhibition of human cytomegalovirus (HCMV) DNA replication. This new scaffold may represent a potential tool useful for the development of effective anti-HAdV drugs.
Collapse
Affiliation(s)
- Sarah Mazzotta
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Seville, Profesor García González 2, E-41071 Seville, Spain
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Cosenza, Italy
| | - Judith Berastegui-Cabrera
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E41013 Seville, Spain
| | - Gabriele Carullo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Cosenza, Italy
- Department of Biotechnology, Chemistry and Pharmacy, DoE 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Margarita Vega-Holm
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Seville, Profesor García González 2, E-41071 Seville, Spain
| | - Marta Carretero-Ledesma
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E41013 Seville, Spain
| | - Lara Mendolia
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Seville, Profesor García González 2, E-41071 Seville, Spain
| | - Francesca Aiello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Cosenza, Italy
| | - Fernando Iglesias-Guerra
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Seville, Profesor García González 2, E-41071 Seville, Spain
| | - Jerónimo Pachón
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E41013 Seville, Spain
- Department of Medicine, University of Seville, E-41009 Seville, Spain
| | - José Manuel Vega-Pérez
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Seville, Profesor García González 2, E-41071 Seville, Spain
| | - Javier Sánchez-Céspedes
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E41013 Seville, Spain
| |
Collapse
|
16
|
Dodge MJ, MacNeil KM, Tessier TM, Weinberg JB, Mymryk JS. Emerging antiviral therapeutics for human adenovirus infection: Recent developments and novel strategies. Antiviral Res 2021; 188:105034. [PMID: 33577808 DOI: 10.1016/j.antiviral.2021.105034] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/11/2022]
Abstract
Human adenoviruses (HAdV) are ubiquitous human pathogens that cause a significant burden of respiratory, ocular, and gastrointestinal illnesses. Although HAdV infections are generally self-limiting, pediatric and immunocompromised individuals are at particular risk for developing severe disease. Currently, no approved antiviral therapies specific to HAdV exist. Recent outbreaks underscore the need for effective antiviral agents to treat life-threatening infections. In this review we will focus on recent developments in search of potential therapeutic agents for controlling HAdV infections, with a focus on those targeting post-entry stages of the virus replicative cycle.
Collapse
Affiliation(s)
- Mackenzie J Dodge
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Katelyn M MacNeil
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Tanner M Tessier
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Jason B Weinberg
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Joe S Mymryk
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada; Department of Otolaryngology, Head & Neck Surgery, The University of Western Ontario, London, ON, Canada; Department of Oncology, The University of Western Ontario, London, ON, Canada; London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
17
|
Xu J, Berastegui-Cabrera J, Carretero-Ledesma M, Chen H, Xue Y, Wold EA, Pachón J, Zhou J, Sánchez-Céspedes J. Discovery of a Small Molecule Inhibitor of Human Adenovirus Capable of Preventing Escape from the Endosome. Int J Mol Sci 2021; 22:ijms22041617. [PMID: 33562748 PMCID: PMC7915867 DOI: 10.3390/ijms22041617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/26/2021] [Accepted: 02/02/2021] [Indexed: 12/16/2022] Open
Abstract
Human adenoviruses (HAdVs) display a wide range of tissue tropism and can cause an array of symptoms from mild respiratory illnesses to disseminated and life-threatening infections in immunocompromised individuals. However, no antiviral drug has been approved specifically for the treatment of HAdV infections. Herein, we report our continued efforts to optimize salicylamide derivatives and discover compound 16 (JMX0493) as a potent inhibitor of HAdV infection. Compound 16 displays submicromolar IC50 values, a higher selectivity index (SI > 100) and 2.5-fold virus yield reduction compared to our hit compound niclosamide. Moreover, unlike niclosamide, our mechanistic studies suggest that the antiviral activity of compound 16 against HAdV is achieved through the inhibition of viral particle escape from the endosome, which bars subsequent uncoating and the presentation of lytic protein VI.
Collapse
Affiliation(s)
- Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA; (J.X.); (H.C.); (Y.X.); (E.A.W.)
| | - Judith Berastegui-Cabrera
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío, CSIC, University of Seville, E41013 Seville, Spain; (J.B.-C.); (M.C.-L.); (J.P.)
| | - Marta Carretero-Ledesma
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío, CSIC, University of Seville, E41013 Seville, Spain; (J.B.-C.); (M.C.-L.); (J.P.)
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA; (J.X.); (H.C.); (Y.X.); (E.A.W.)
| | - Yu Xue
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA; (J.X.); (H.C.); (Y.X.); (E.A.W.)
| | - Eric A. Wold
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA; (J.X.); (H.C.); (Y.X.); (E.A.W.)
| | - Jerónimo Pachón
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío, CSIC, University of Seville, E41013 Seville, Spain; (J.B.-C.); (M.C.-L.); (J.P.)
- Department of Medicine, University of Seville, E-41009 Seville, Spain
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA; (J.X.); (H.C.); (Y.X.); (E.A.W.)
- Correspondence: (J.Z.); (J.S.-C.); Tel.: +(1)-409-772-9748 (J.Z.); +(34)-955-923-100 (J.S.-C.)
| | - Javier Sánchez-Céspedes
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío, CSIC, University of Seville, E41013 Seville, Spain; (J.B.-C.); (M.C.-L.); (J.P.)
- Correspondence: (J.Z.); (J.S.-C.); Tel.: +(1)-409-772-9748 (J.Z.); +(34)-955-923-100 (J.S.-C.)
| |
Collapse
|
18
|
Xu J, Berastegui-Cabrera J, Ye N, Carretero-Ledesma M, Pachón-Díaz J, Chen H, Pachón-Ibáñez ME, Sánchez-Céspedes J, Zhou J. Discovery of Novel Substituted N-(4-Amino-2-chlorophenyl)-5-chloro-2-hydroxybenzamide Analogues as Potent Human Adenovirus Inhibitors. J Med Chem 2020; 63:12830-12852. [PMID: 33112138 DOI: 10.1021/acs.jmedchem.0c01226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An effective therapy for human adenovirus (HAdV) infections in immunocompromised patients and healthy individuals with community-acquired pneumonia remains an unmet medical need. We herein reported a series of novel substituted N-(4-amino-2-chlorophenyl)-5-chloro-2-hydroxybenzamide analogues as potent HAdV inhibitors. Compounds 6, 15, 29, 40, 43, 46, 47, and 54 exhibited increased selectivity indexes (SI > 100) compared to the lead compound niclosamide, while maintaining sub-micromolar to low micromolar potency against HAdV. The preliminary mechanistic studies indicated that compounds 6 and 43 possibly target the HAdV DNA replication process, while compounds 46 and 47 suppress later steps of HAdV life cycle. Notably, among these derivatives, compound 15 showed improved anti-HAdV activity (IC50 = 0.27 μM), significantly decreased cytotoxicity (CC50 = 156.8 μM), and low in vivo toxicity (maximum tolerated dose = 150 mg/kg in hamster) as compared with niclosamide, supporting its further in vivo efficacy studies for the treatment of HAdV infections.
Collapse
Affiliation(s)
- Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Judith Berastegui-Cabrera
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E-41013 Seville, Spain
| | - Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Marta Carretero-Ledesma
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E-41013 Seville, Spain
| | - Jerónimo Pachón-Díaz
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E-41013 Seville, Spain.,Department of Medicine, University of Seville, E-41009 Seville, Spain
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Maria Eugenia Pachón-Ibáñez
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E-41013 Seville, Spain
| | - Javier Sánchez-Céspedes
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E-41013 Seville, Spain
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
19
|
Toth K, Hussein ITM, Tollefson AE, Ying B, Spencer JF, Eagar J, James SH, Prichard MN, Wold WSM, Bowlin TL. Filociclovir Is a Potent In Vitro and In Vivo Inhibitor of Human Adenoviruses. Antimicrob Agents Chemother 2020; 64:e01299-20. [PMID: 32816736 PMCID: PMC7577159 DOI: 10.1128/aac.01299-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/13/2020] [Indexed: 12/24/2022] Open
Abstract
Human adenovirus (HAdV) infection is common in the general population and can cause a range of clinical manifestations, among which pneumonia and keratoconjunctivitis are the most common. Although HAdV infections are mostly self-limiting, infections in immunocompromised individuals can be severe. No antiviral drug has been approved for treating adenoviruses. Filociclovir (FCV) is a nucleoside analogue which has successfully completed phase I human clinical safety studies and is now being developed for treatment of human cytomegalovirus (HCMV)-related disease in immunocompromised patients. In this report, we show that FCV is a potent broad-spectrum inhibitor of HAdV types 4 to 8, with 50% effective concentrations (EC50s) ranging between 1.24 and 3.6 μM and a 50% cytotoxic concentration (CC50) of 100 to 150 μM in human foreskin fibroblasts (HFFs). We also show that the prophylactic oral administration of FCV (10 mg/kg of body weight) 1 day prior to virus challenge and then daily for 14 days to immunosuppressed Syrian hamsters infected intravenously with HAdV6 was sufficient to prevent morbidity and mortality. FCV also mitigated tissue damage and inhibited virus replication in the liver. The 10-mg/kg dose had similar effects even when the treatment was started on day 4 after virus challenge. Furthermore, FCV administered at the same dose after intranasal challenge with HAdV6 partially mitigated body weight loss but significantly reduced pathology and virus replication in the lung. These findings suggest that FCV could potentially be developed as a pan-adenoviral inhibitor.
Collapse
Affiliation(s)
- Karoly Toth
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | | | - Ann E Tollefson
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Baoling Ying
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Jacqueline F Spencer
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Jessica Eagar
- Department of Pediatrics, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Scott H James
- Department of Pediatrics, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mark N Prichard
- Department of Pediatrics, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - William S M Wold
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
20
|
Jennings MR, Parks RJ. Antiviral Effects of Curcumin on Adenovirus Replication. Microorganisms 2020; 8:microorganisms8101524. [PMID: 33020422 PMCID: PMC7599685 DOI: 10.3390/microorganisms8101524] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 12/15/2022] Open
Abstract
Human adenovirus (HAdV) is a common pathogen that can cause severe morbidity and mortality in certain populations, including pediatric, geriatric, and immunocompromised patients. Unfortunately, there are no approved therapeutics to combat HAdV infections. Curcumin, the primary curcuminoid compound found in turmeric spice, has shown broad activity as an antimicrobial agent, limiting the replication of many different bacteria and viruses. In this study, we evaluated curcumin as an anti-HAdV agent. Treatment of cells in culture with curcumin reduced HAdV replication, gene expression, and virus yield, at concentrations of curcumin that had little effect on cell viability. Thus, curcumin represents a promising class of compounds for further study as potential therapeutics to combat HAdV infection.
Collapse
Affiliation(s)
- Morgan R. Jennings
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada;
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Robin J. Parks
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada;
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Department of Medicine, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
- Correspondence: ; Tel.: +1-613-737-8123
| |
Collapse
|
21
|
Adenovirus infection and disease in recipients of hematopoietic cell transplantation. Curr Opin Infect Dis 2020; 32:591-600. [PMID: 31567568 DOI: 10.1097/qco.0000000000000605] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW To provide an update on risk factors associated with adenovirus (ADV) infection in patients after hematopoietic cell transplant (HCT) and on options for ADV monitoring and treatment in the setting of HCT. RECENT FINDINGS Among patients undergoing HCT, ADV infection continues to be more common amongst those receiving a T-cell-depleted or graft other than from a matched-related donor. Among children undergoing HCT, reactivation in the gastrointestinal tract appears to be the most common source, and the virus is detectable by quantitative PCR in the stool before it is detectable in the blood. Thus, screening for the virus in the stool of these children may allow for preemptive therapy to reduce mortality. Brincidofovir, although still not approved by any regulatory agency, remains a potential agent for preemptive therapy and for salvage in cases not responding to cidofovir. Rapidly generated off-the-shelf virus-specific T cells may facilitate adoptive cell therapy in populations with a special need and previously not eligible for adoptive cell therapy, such as cord blood recipients. SUMMARY ADV infection continues to adversely affect survival in HCT recipients. Screening stool in children and preemptive therapy may reduce mortality. Brincidofovir and adoptive T-cell therapy remain potential options for treatment.
Collapse
|
22
|
Xu J, Berastegui-Cabrera J, Chen H, Pachón J, Zhou J, Sánchez-Céspedes J. Structure-Activity Relationship Studies on Diversified Salicylamide Derivatives as Potent Inhibitors of Human Adenovirus Infection. J Med Chem 2020; 63:3142-3160. [PMID: 32045239 DOI: 10.1021/acs.jmedchem.9b01950] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The effective treatment of adenovirus (HAdV) infections in immunocompromised patients still poses great challenges. Herein, we reported our continued efforts to optimize a series of salicylamide derivatives as potent inhibitors of HAdV infection. Of these, nine compounds (11, 13, 14, 17, 20, 58, 60, 62, and 70) showed significantly improved anti-HAdV activities with nanomolar to submicromolar IC50 values and high selectivity indexes (SI > 100), indicating better safety windows, compared to those of the lead compound niclosamide. Our mechanistic assays suggest that compounds 13, 62, and 70 exert their activities in the HAdV entry pathway, while compounds 14 and 60 likely target the HAdV DNA replication, and 11, 17, 20, and 58 inhibit later steps after DNA replication. Given the broad anti-viral activity profile of niclosamide, these derivatives may also offer therapeutic potential for other viral infections.
Collapse
Affiliation(s)
- Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Judith Berastegui-Cabrera
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E41013 Seville, Spain
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jerónimo Pachón
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E41013 Seville, Spain.,Department of Medicine, University of Seville, E-41009 Seville, Spain
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Javier Sánchez-Céspedes
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E41013 Seville, Spain
| |
Collapse
|
23
|
Alcamo AM, Wolf MS, Alessi LJ, Chong HJ, Green M, Williams JV, Simon DW. Successful Use of Cidofovir in an Immunocompetent Child With Severe Adenoviral Sepsis. Pediatrics 2020; 145:peds.2019-1632. [PMID: 31826930 PMCID: PMC6939840 DOI: 10.1542/peds.2019-1632] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2019] [Indexed: 01/22/2023] Open
Abstract
Adenovirus infection is common in childhood and is generally associated with self-limited disease. Cidofovir, a viral DNA polymerase inhibitor, is used to treat adenovirus infection in select populations but is not often recommended for immunocompetent patients because of limited antiviral activity and nephrotoxicity. Here, we report a case of fulminant adenovirus infection associated with lymphopenia and multiple organ failure requiring extracorporeal membrane oxygenation support in a previously healthy child. After 1 week of supportive therapy, the patient had persistent organ failure and continued to have adenoviremia of >560 000 copies per mL. Weekly doses of cidofovir with concurrent probenecid for renal protection was initiated. Adenovirus blood load declined after the first cidofovir dose, becoming undetectable after 3 doses. The patient was successfully decannulated from extracorporeal membrane oxygenation, extubated, and eventually discharged at his functional baseline without need for ongoing respiratory support. Lymphopenia improved after viremia resolved, and a subsequent immunologic workup revealed no evidence of primary immunodeficiency. The viral isolate was genotyped as adenovirus type 7. This case reveals the successful use of cidofovir for management of severe adenovirus infection in a previously healthy child. To date, there are no universally accepted recommendations for the use of cidofovir in this population. Further study is warranted to determine the potential role of cidofovir in treating severe adenovirus infections in immunocompetent children.
Collapse
Affiliation(s)
- Alicia M. Alcamo
- Departments of Critical Care Medicine and,Contributed equally as co-first authors
| | - Michael S. Wolf
- Departments of Critical Care Medicine and,Contributed equally as co-first authors
| | | | - Hey J. Chong
- Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael Green
- Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John V. Williams
- Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | |
Collapse
|
24
|
Agostini ML, Pruijssers AJ, Chappell JD, Gribble J, Lu X, Andres EL, Bluemling GR, Lockwood MA, Sheahan TP, Sims AC, Natchus MG, Saindane M, Kolykhalov AA, Painter GR, Baric RS, Denison MR. Small-Molecule Antiviral β-d- N4-Hydroxycytidine Inhibits a Proofreading-Intact Coronavirus with a High Genetic Barrier to Resistance. J Virol 2019; 93:e01348-19. [PMID: 31578288 PMCID: PMC6880162 DOI: 10.1128/jvi.01348-19] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 09/24/2019] [Indexed: 12/22/2022] Open
Abstract
Coronaviruses (CoVs) have emerged from animal reservoirs to cause severe and lethal disease in humans, but there are currently no FDA-approved antivirals to treat the infections. One class of antiviral compounds, nucleoside analogues, mimics naturally occurring nucleosides to inhibit viral replication. While these compounds have been successful therapeutics for several viral infections, mutagenic nucleoside analogues, such as ribavirin and 5-fluorouracil, have been ineffective at inhibiting CoVs. This has been attributed to the proofreading activity of the viral 3'-5' exoribonuclease (ExoN). β-d-N4-Hydroxycytidine (NHC) (EIDD-1931; Emory Institute for Drug Development) has recently been reported to inhibit multiple viruses. Here, we demonstrate that NHC inhibits both murine hepatitis virus (MHV) (50% effective concentration [EC50] = 0.17 μM) and Middle East respiratory syndrome CoV (MERS-CoV) (EC50 = 0.56 μM) with minimal cytotoxicity. NHC inhibited MHV lacking ExoN proofreading activity similarly to wild-type (WT) MHV, suggesting an ability to evade or overcome ExoN activity. NHC inhibited MHV only when added early during infection, decreased viral specific infectivity, and increased the number and proportion of G:A and C:U transition mutations present after a single infection. Low-level NHC resistance was difficult to achieve and was associated with multiple transition mutations across the genome in both MHV and MERS-CoV. These results point to a virus-mutagenic mechanism of NHC inhibition in CoVs and indicate a high genetic barrier to NHC resistance. Together, the data support further development of NHC for treatment of CoVs and suggest a novel mechanism of NHC interaction with the CoV replication complex that may shed light on critical aspects of replication.IMPORTANCE The emergence of coronaviruses (CoVs) into human populations from animal reservoirs has demonstrated their epidemic capability, pandemic potential, and ability to cause severe disease. However, no antivirals have been approved to treat these infections. Here, we demonstrate the potent antiviral activity of a broad-spectrum ribonucleoside analogue, β-d-N4-hydroxycytidine (NHC), against two divergent CoVs. Viral proofreading activity does not markedly impact sensitivity to NHC inhibition, suggesting a novel interaction between a nucleoside analogue inhibitor and the CoV replicase. Further, passage in the presence of NHC generates only low-level resistance, likely due to the accumulation of multiple potentially deleterious transition mutations. Together, these data support a mutagenic mechanism of inhibition by NHC and further support the development of NHC for treatment of CoV infections.
Collapse
Affiliation(s)
- Maria L Agostini
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Andrea J Pruijssers
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - James D Chappell
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jennifer Gribble
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Xiaotao Lu
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Erica L Andres
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Gregory R Bluemling
- Emory Institute for Drug Development, Emory University, Atlanta, Georgia, USA
| | - Mark A Lockwood
- Emory Institute for Drug Development, Emory University, Atlanta, Georgia, USA
| | - Timothy P Sheahan
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Amy C Sims
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael G Natchus
- Emory Institute for Drug Development, Emory University, Atlanta, Georgia, USA
| | - Manohar Saindane
- Emory Institute for Drug Development, Emory University, Atlanta, Georgia, USA
| | | | - George R Painter
- Emory Institute for Drug Development, Emory University, Atlanta, Georgia, USA
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mark R Denison
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
25
|
Wold WSM, Tollefson AE, Ying B, Spencer JF, Toth K. Drug development against human adenoviruses and its advancement by Syrian hamster models. FEMS Microbiol Rev 2019; 43:380-388. [PMID: 30916746 DOI: 10.1093/femsre/fuz008] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 03/25/2019] [Indexed: 02/02/2023] Open
Abstract
The symptoms of human adenovirus infections are generally mild and self-limiting. However, these infections have been gaining importance in recent years because of a growing number of immunocompromised patients. Solid organ and hematopoietic stem cell transplant patients are subjected to severe immunosuppressive regimes and cannot efficaciously eliminate virus infections. In these patients, adenovirus infections can develop into deadly multi-organ disseminated disease. Presently, in the absence of approved therapies, physicians rely on drugs developed for other purposes to treat adenovirus infections. As there is a need for anti-adenoviral therapies, researchers have been developing new agents and repurposing existing ones to treat adenovirus infections. There are several small molecule drugs that are being tested for their efficacy against human adenoviruses; some of these have reached clinical trials, while others are still in the preclinical phase. Besides these compounds, research on immunotherapy against adenoviral infection has made significant progress, promising another modality for treatment. The availability of an animal model confirmed the activity of some drugs already in clinical use while proving that others are inactive. This led to the identification of several lead compounds that await further development. In the present article, we review the current status of anti-adenoviral therapies and their advancement by in vivo studies in the Syrian hamster model.
Collapse
Affiliation(s)
- William S M Wold
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO, USA
| | - Ann E Tollefson
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO, USA
| | - Baoling Ying
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO, USA
| | - Jacqueline F Spencer
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO, USA
| | - Karoly Toth
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO, USA
| |
Collapse
|