1
|
Gege C, Kleymann G. Helicase-primase inhibitors for the treatment of herpes simplex virus infections - patent evaluation of WO2023/225162 from Gilead Sciences Inc. Expert Opin Ther Pat 2024; 34:863-872. [PMID: 39262042 DOI: 10.1080/13543776.2024.2403618] [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: 06/14/2024] [Revised: 08/09/2024] [Accepted: 08/22/2024] [Indexed: 09/13/2024]
Abstract
Helicase-primase is an interesting target for small-molecule therapy of herpes simplex virus (HSV) infections. With amenamevir already approved for varicella-zoster virus and herpes simplex in Japan and with pritelivir's granted breakthrough therapy designation for the treatment of acyclovir-resistant HSV infections in immunocompromised patients, the target has sparked interest in helicase-primase inhibitors (HPIs). Here, we analyze the first patent application from Gilead in this field, which pursued a me-too approach combining elements from an old Bayer together with a recent Medshine HPI application (which covers the Phaeno Therapeutics drug candidate HN0037). The asset was contributed to Assembly Biosciences, where it is under development as ABI-1179 at the investigational new drug (IND) enabling stage for high-recurrence genital herpes. A structure proposal for indolinoyl derivative ABI-1179 is presented, showing its potential opportunities and limitations compared to other HPIs.
Collapse
|
2
|
Deng H, Cao H, Wang Y, Li J, Dai J, Li LF, Qiu HJ, Li S. Viral replication organelles: the highly complex and programmed replication machinery. Front Microbiol 2024; 15:1450060. [PMID: 39144209 PMCID: PMC11322364 DOI: 10.3389/fmicb.2024.1450060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Viral infections usually induce the rearrangement of cellular cytoskeletal proteins and organelle membrane structures, thus creating independent compartments [termed replication organelles (ROs)] to facilitate viral genome replication. Within the ROs, viral replicases, including polymerases, helicases, and ligases, play functional roles during viral replication. These viral replicases are pivotal in the virus life cycle, and numerous studies have demonstrated that the viral replicases could be the potential targets for drugs development. Here, we summarize primarily the key replicases within viral ROs and emphasize the advancements of antiviral drugs targeting crucial viral replicases, providing novel insights into the future development of antiviral strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-reference Laboratory, National High Containment Facilities for Animal Diseases Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Su Li
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-reference Laboratory, National High Containment Facilities for Animal Diseases Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
3
|
Gege C, Kleymann G. Replacement of sulfonamide by sulfoximine within a helicase-primase inhibitor with restricted flexibility. Bioorg Med Chem Lett 2024; 106:129761. [PMID: 38642810 DOI: 10.1016/j.bmcl.2024.129761] [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: 01/31/2024] [Revised: 03/14/2024] [Accepted: 04/18/2024] [Indexed: 04/22/2024]
Abstract
Helicase-primase is an interesting target for the therapy of herpes simplex virus (HSV) infections. Since amenamevir is already approved for varicella-zoster virus (VZV) and HSV in Japan and pritelivir has received breakthrough therapy status for the treatment of acyclovir-resistant HSV infections in immunocompromised patients, the target has sparked interest in me-too approaches. Here, we describe the attempt to improve nervous tissue penetration in Phaeno Therapeutics drug candidate HN0037 to target the latent reservoir of HSV by installing less polar moieties, mainly a difluorophenyl instead of a pyridyl group, and replacing the primary sulfonamide with a methyl sulfoximine moiety. However, all obtained stereoisomers exhibited a weaker inhibitory activity on HSV-1 and HSV-2.
Collapse
Affiliation(s)
- Christian Gege
- Innovative Molecules GmbH, Lipowsky Str. 10, 81373 Munich, Germany.
| | - Gerald Kleymann
- Innovative Molecules GmbH, Lipowsky Str. 10, 81373 Munich, Germany
| |
Collapse
|
4
|
Lorenzi A, Legeai F, Jouan V, Girard PA, Strand MR, Ravallec M, Eychenne M, Bretaudeau A, Robin S, Rochefort J, Villegas M, Burke GR, Rebollo R, Nègre N, Volkoff AN. Identification of a viral gene essential for the genome replication of a domesticated endogenous virus in ichneumonid parasitoid wasps. PLoS Pathog 2024; 20:e1011980. [PMID: 38662774 DOI: 10.1371/journal.ppat.1011980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/07/2024] [Accepted: 03/22/2024] [Indexed: 05/08/2024] Open
Abstract
Thousands of endoparasitoid wasp species in the families Braconidae and Ichneumonidae harbor "domesticated endogenous viruses" (DEVs) in their genomes. This study focuses on ichneumonid DEVs, named ichnoviruses (IVs). Large quantities of DNA-containing IV virions are produced in ovary calyx cells during the pupal and adult stages of female wasps. Females parasitize host insects by injecting eggs and virions into the body cavity. After injection, virions rapidly infect host cells which is followed by expression of IV genes that promote the successful development of wasp offspring. IV genomes consist of two components: proviral segment loci that serve as templates for circular dsDNAs that are packaged into capsids, and genes from an ancestral virus that produce virions. In this study, we generated a chromosome-scale genome assembly for Hyposoter didymator that harbors H. didymator ichnovirus (HdIV). We identified a total of 67 HdIV loci that are amplified in calyx cells during the wasp pupal stage. We then focused on an HdIV gene, U16, which is transcribed in calyx cells during the initial stages of replication. Sequence analysis indicated that U16 contains a conserved domain in primases from select other viruses. Knockdown of U16 by RNA interference inhibited virion morphogenesis in calyx cells. Genome-wide analysis indicated U16 knockdown also inhibited amplification of HdIV loci in calyx cells. Altogether, our results identified several previously unknown HdIV loci, demonstrated that all HdIV loci are amplified in calyx cells during the pupal stage, and showed that U16 is required for amplification and virion morphogenesis.
Collapse
Affiliation(s)
- Ange Lorenzi
- DGIMI, Univ Montpellier, INRAE, Montpellier, France
- Department of Entomology, University of Georgia, Athens, Georgia, United States of America
| | - Fabrice Legeai
- INRAE, UMR Institut de Génétique, Environnement et Protection des Plantes (IGEPP), BioInformatics Platform for Agroecosystems Arthropods (BIPAA), Campus Beaulieu, Rennes, France
- INRIA, IRISA, GenOuest Core Facility, Campus de Beaulieu, Rennes, France
| | | | | | - Michael R Strand
- Department of Entomology, University of Georgia, Athens, Georgia, United States of America
| | | | | | - Anthony Bretaudeau
- INRAE, UMR Institut de Génétique, Environnement et Protection des Plantes (IGEPP), BioInformatics Platform for Agroecosystems Arthropods (BIPAA), Campus Beaulieu, Rennes, France
- INRIA, IRISA, GenOuest Core Facility, Campus de Beaulieu, Rennes, France
| | - Stéphanie Robin
- INRAE, UMR Institut de Génétique, Environnement et Protection des Plantes (IGEPP), BioInformatics Platform for Agroecosystems Arthropods (BIPAA), Campus Beaulieu, Rennes, France
- INRIA, IRISA, GenOuest Core Facility, Campus de Beaulieu, Rennes, France
| | | | | | - Gaelen R Burke
- Department of Entomology, University of Georgia, Athens, Georgia, United States of America
| | - Rita Rebollo
- INRAE, INSA Lyon, BF2I, UMR203, Villeurbanne, France
| | | | | |
Collapse
|
5
|
Mohammed HS, Taha EFS, Mahrous FS, Sabour R, Abdel-Aziz MM, Ismail LD. Antimicrobial and antiviral evaluation of compounds from Holoptelea integrifolia: in silico supported in vitro study. RSC Adv 2023; 13:32473-32486. [PMID: 37928846 PMCID: PMC10624013 DOI: 10.1039/d3ra05978b] [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: 09/02/2023] [Accepted: 10/29/2023] [Indexed: 11/07/2023] Open
Abstract
Holoptelea integrifolia, also known as the Indian Elm Tree, has been used in Ayurvedic medicine for its medicinal properties. In this study, two biologically active metabolites, 5(6) dihydrostigmast 22en 3-O-β-glucoside (DHS) and 1-O-eicosanoyl glycerol-2'-O-β-galactouronic (EGG), were isolated for the first time from the n-butanol fraction of H. integrifolia using a chromatographic technique and identified by NMR, and HRESI-MS. The antiviral and multidrug-resistant activities of these metabolites were evaluated as well as the n-butanol fraction. The n-butanol fraction of H. integrifolia exhibited weak antiviral effects, but DHS and EGG demonstrated significant antiviral activity against herpes simplex type-1 (HSV-1) and Coxsackie (CoxB4) viruses. Both metabolites showed lower IC50 values than the standard antiviral drug acyclovir, indicating their potency in inhibiting viral replication. EGG showed potent antiviral activity with minimal cytotoxicity at the highest concentration tested, presenting a selectivity index (SI) of 18.18 and 15.58 against HSV-1 and CoxB4 viruses, respectively. A preliminary assessment of the antibacterial activity of the n-butanol fraction and metabolites revealed that DHS had the highest inhibitory potency against drug-resistant strains, including MRSA and Carbapenem-resistant Klebsiella pneumonia. It also exhibited significant inhibitions against Fluconazole-resistant Candida albicans and ESBL - Escherichia coli. DHS displayed the lowest minimum inhibitory concentration (MIC) values, indicating its superiority as an antibacterial agent compared to EGG and the n-butanol fraction. Molecular docking analysis confirmed the antiviral and antibacterial actions of DHS and EGG by demonstrating their strong binding.
Collapse
Affiliation(s)
- Hala Sh Mohammed
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Girls), Al-Azhar University Cairo (11311) Egypt
| | - Eman F S Taha
- Department of Health Radiation Research, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA) Cairo Egypt
| | - Fatma S Mahrous
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Girls), Al-Azhar University Cairo (11311) Egypt
| | - Rehab Sabour
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University Cairo Egypt
| | - Marwa M Abdel-Aziz
- The Regional Centre for Mycology and Biotechnology, Al-Azhar University Cairo Egypt
| | - Lotfy D Ismail
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Boys), Al-Azhar University Cairo Egypt
| |
Collapse
|
6
|
Malahe SRK, van Kampen JJA, Manintveld OC, Hoek RAS, den Hoed CM, Baan CC, Kho MML, Verjans GMGM. Current Perspectives on the Management of Herpesvirus Infections in Solid Organ Transplant Recipients. Viruses 2023; 15:1595. [PMID: 37515280 PMCID: PMC10383436 DOI: 10.3390/v15071595] [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: 06/16/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Solid organ transplant recipients (SOTRs) are at high risk of human herpesvirus (HHV)-related morbidity and mortality due to the use of immunosuppressive therapy. We aim to increase awareness and understanding of HHV disease burden in SOTRs by providing an overview of current prevention and management strategies as described in the literature and guidelines. We discuss challenges in both prevention and treatment as well as future perspectives.
Collapse
Affiliation(s)
- S Reshwan K Malahe
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Jeroen J A van Kampen
- Department of Viroscience, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Olivier C Manintveld
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Cardiology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Rogier A S Hoek
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Caroline M den Hoed
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Marcia M L Kho
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Georges M G M Verjans
- Department of Viroscience, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- HerpeslabNL, Department of Viroscience, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
7
|
Gege C, Kleymann G. Helicase-primase inhibitors from Medshine Discovery Inc. (WO2018/127207 and WO2020/007355) for the treatment of herpes simplex virus infections – structure proposal for Phaeno Therapeutics drug candidate HN0037. Expert Opin Ther Pat 2022; 32:933-937. [DOI: 10.1080/13543776.2022.2113873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Christian Gege
- Innovative Molecules GmbH, Dachauer Str. 65, 80335 München, Germany
| | - Gerald Kleymann
- Innovative Molecules GmbH, Dachauer Str. 65, 80335 München, Germany
| |
Collapse
|
8
|
Zhang X, Hao K, Li S, Meng L, Chen H, Wei F, Yu F, Xu J, Zhao Z. Channel catfish virus ORF25 and ORF63 genes are essential for viral replication in vitro. JOURNAL OF FISH DISEASES 2022; 45:655-666. [PMID: 35176182 DOI: 10.1111/jfd.13591] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 06/14/2023]
Abstract
The channel catfish virus (CCV) is a lethal pathogen to aquatic animals that can provoke severe haemorrhagic disease in juvenile channel catfish. Although the CCV genome has been fully sequenced, the molecular mechanisms of CCV infection and pathogenesis are less well known. Genomic DNA replication is a necessary and key event for the CCV life cycle. In this study, the impacts of the putative helicase and primase encoded by viral ORF25 and ORF63 on CCV genome replication and infection were evaluated in channel catfish ovary (CCO) cells. The results showed that the number of CCV genome copies was decreased significantly in virus-infected CCO cells after knockdown of ORF25 and ORF63 using RNA interference. In contrast, the overexpression of ORF25 and ORF63 led to slight increase in the number of virus genome copies. Consistent with the above results, the present results also showed that the expressions of CCV true-late genes which strictly depend on viral DNA replication, were significantly increased or repressed by overexpression or RNA interference targeting viral ORF25 and ORF63 genes in virus-infected CCO cells. In addition, knockdown of ORF25 and ORF63 remarkably inhibited CCV-induced cytopathic effects and decreased progeny virus titres in CCO cells. Moreover, transmission electron microscopy observation of CCO cells infected with CCV accompanied by siRNA targeting the viral ORF25 and ORF63 genes showed that the number of virus particles was remarkably reduced. Taken together, these results indicated that ORF25 and ORF63 are essential for regulating CCV genome replication and CCV-induced infection. Our findings will provide an understanding of the replication mechanisms of CCV and contribute to the development of antiviral strategies for controlling CCV infection in channel catfish culture.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, China
| | - Kai Hao
- Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, China
| | - Shuxin Li
- Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, China
| | - Lihui Meng
- Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, China
| | - Hongxun Chen
- Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, China
| | - Fucheng Wei
- Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, China
| | - Fei Yu
- Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, China
| | - Jing Xu
- Jiangsu Cangdong Agricultural Development Co., Ltd, Nanjing, China
| | - Zhe Zhao
- Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, China
| |
Collapse
|
9
|
Majewska A, Mlynarczyk-Bonikowska B. 40 Years after the Registration of Acyclovir: Do We Need New Anti-Herpetic Drugs? Int J Mol Sci 2022; 23:ijms23073431. [PMID: 35408788 PMCID: PMC8998721 DOI: 10.3390/ijms23073431] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/10/2022] [Accepted: 03/18/2022] [Indexed: 01/17/2023] Open
Abstract
Herpes simplex virus types 1 and 2 HSV1 and 2, namely varicella-zoster VZV and cytomegalovirus CMV, are among the most common pathogens worldwide. They remain in the host body for life. The course of infection with these viruses is often asymptomatic or mild and self-limiting, but in immunocompromised patients, such as solid organ or bone marrow transplant recipients, the course can be very severe or even life-threatening. Unfortunately, in the latter group, the highest percentage of infections with strains resistant to routinely used drugs is observed. On the other hand, frequent recurrences of genital herpes can be a problem even in people with normal immunity. Genital herpes also increases the risk of acquiring sexually transmitted diseases, including HIV infection and, if present in pregnant women, poses a risk to the fetus and newborn. Even more frequently than herpes simplex, congenital infections can be caused by cytomegalovirus. We present the most important anti-herpesviral agents, the mechanisms of resistance to these drugs, and the associated mutations in the viral genome. Special emphasis was placed on newly introduced drugs such as maribavir and brincidofovir. We also briefly discuss the most promising substances in preclinical testing as well as immunotherapy options and vaccines currently in use and under investigation.
Collapse
Affiliation(s)
- Anna Majewska
- Department of Medical Microbiology, Medical University of Warsaw, Chałubińskiego 5, 02-004 Warsaw, Poland;
| | - Beata Mlynarczyk-Bonikowska
- Department of Dermatology, Immunodermatology and Venereology, Medical University of Warsaw, Koszykowa 82a, 02-008 Warsaw, Poland
- Correspondence: ; Tel.: +48-225021313
| |
Collapse
|
10
|
Bermek O, Williams RS. The three-component helicase/primase complex of herpes simplex virus-1. Open Biol 2021; 11:210011. [PMID: 34102080 PMCID: PMC8187027 DOI: 10.1098/rsob.210011] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is one of the nine herpesviruses that infect humans. HSV-1 encodes seven proteins to replicate its genome in the hijacked human cell. Among these are the herpes virus DNA helicase and primase that are essential components of its replication machinery. In the HSV-1 replisome, the helicase-primase complex is composed of three components including UL5 (helicase), UL52 (primase) and UL8 (non-catalytic subunit). UL5 and UL52 subunits are functionally interdependent, and the UL8 component is required for the coordination of UL5 and UL52 activities proceeding in opposite directions with respect to the viral replication fork. Anti-viral compounds currently under development target the functions of UL5 and UL52. Here, we review the structural and functional properties of the UL5/UL8/UL52 complex and highlight the gaps in knowledge to be filled to facilitate molecular characterization of the structure and function of the helicase-primase complex for development of alternative anti-viral treatments.
Collapse
Affiliation(s)
- Oya Bermek
- Genome Integrity and Structural Biology Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - R Scott Williams
- Genome Integrity and Structural Biology Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
11
|
Spratt AN, Gallazzi F, Quinn TP, Lorson CL, Sönnerborg A, Singh K. Coronavirus helicases: attractive and unique targets of antiviral drug-development and therapeutic patents. Expert Opin Ther Pat 2021; 31:339-350. [PMID: 33593200 PMCID: PMC8074651 DOI: 10.1080/13543776.2021.1884224] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Introduction: Coronaviruses encode a helicase that is essential for viral replication and represents an excellent antiviral target. However, only a few coronavirus helicase inhibitors have been patented. These patents include drug-like compound SSYA10-001, aryl diketo acids (ADK), and dihydroxychromones. Additionally, adamantane-derived bananins, natural flavonoids, one acrylamide derivative [(E)-3-(furan-2-yl)-N-(4-sulfamoylphenyl)acrylamide], a purine derivative (7-ethyl-8-mercapto-3-methyl-3,7-dihydro-1 H-purine-2,6-dione), and a few bismuth complexes. The IC50 of patented inhibitors ranges between 0.82 μM and 8.95 μM, depending upon the assays used. Considering the urgency of clinical interventions against Coronavirus Disease-19 (COVID-19), it is important to consider developing antiviral portfolios consisting of small molecules. Areas covered: This review examines coronavirus helicases as antiviral targets, and the potential of previously patented and experimental compounds to inhibit the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) helicase. Expert opinion: Small molecule coronavirus helicase inhibitors represent attractive pharmacological modalities for the treatment of coronaviruses such as SARS-CoV and SARS-CoV-2. Rightfully so, the current emphasis is focused upon the development of vaccines. However, vaccines may not work for everyone and broad-based adoption of vaccinations is an increasingly challenging societal endeavor. Therefore, it is important to develop additional pharmacological antivirals against the highly conserved coronavirus helicases to broadly protect against this and subsequent coronavirus epidemics.
Collapse
Affiliation(s)
- Austin N Spratt
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Fabio Gallazzi
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Chemistry, University of Missouri, Columbia, MO, USA
| | - Thomas P Quinn
- cDepartment of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Christian L Lorson
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,dDepartment of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Anders Sönnerborg
- eDivision of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Huddinge, Stockholm, Sweden.,fDepartment of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Kamal Singh
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA.,Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Huddinge, Stockholm, Sweden.,gSanctum Therapeutics Corporation, Sunnyvale, CA, USA
| |
Collapse
|
12
|
Didychuk AL, Gates SN, Gardner MR, Strong LM, Martin A, Glaunsinger BA. A pentameric protein ring with novel architecture is required for herpesviral packaging. eLife 2021; 10:e62261. [PMID: 33554858 PMCID: PMC7889075 DOI: 10.7554/elife.62261] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
Genome packaging in large double-stranded DNA viruses requires a powerful molecular motor to force the viral genome into nascent capsids, which involves essential accessory factors that are poorly understood. Here, we present structures of two such accessory factors from the oncogenic herpesviruses Kaposi's sarcoma-associated herpesvirus (KSHV; ORF68) and Epstein-Barr virus (EBV; BFLF1). These homologous proteins form highly similar homopentameric rings with a positively charged central channel that binds double-stranded DNA. Mutation of individual positively charged residues within but not outside the channel ablates DNA binding, and in the context of KSHV infection, these mutants fail to package the viral genome or produce progeny virions. Thus, we propose a model in which ORF68 facilitates the transfer of newly replicated viral genomes to the packaging motor.
Collapse
Affiliation(s)
- Allison L Didychuk
- Department of Plant and Microbial Biology, University of California, BerkeleyBerkeleyUnited States
| | - Stephanie N Gates
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
- California Institute for Quantitative Biosciences, University of California, BerkeleyBerkeleyUnited States
- Howard Hughes Medical Institute, University of California, BerkeleyBerkeleyUnited States
| | - Matthew R Gardner
- Department of Plant and Microbial Biology, University of California, BerkeleyBerkeleyUnited States
| | - Lisa M Strong
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Andreas Martin
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
- California Institute for Quantitative Biosciences, University of California, BerkeleyBerkeleyUnited States
- Howard Hughes Medical Institute, University of California, BerkeleyBerkeleyUnited States
| | - Britt A Glaunsinger
- Department of Plant and Microbial Biology, University of California, BerkeleyBerkeleyUnited States
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
- California Institute for Quantitative Biosciences, University of California, BerkeleyBerkeleyUnited States
- Howard Hughes Medical Institute, University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
13
|
Mello CP, Quirico-Santos T, Amorim LF, Silva VG, Fragel LM, Bloom DC, Paixão IP. Perillyl alcohol and perillic acid exert efficient action upon HSV-1 maturation and release of infective virus. Antivir Ther 2019; 25:1-11. [PMID: 31099756 DOI: 10.3851/imp3315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Infection by herpes simplex type-1 virus (HSV-1) causes several pathological processes, including cutaneous, oral and genital infections, fatal encephalitis and cognitive dysfunction due to grey matter loss. Acyclovir is the reference compound used as HSV-1 antiviral therapy. However, with the emergence of HSV-resistant strains to current antiviral drugs, development of new antiviral agents with distinct modes of action is urgently needed. METHODS In this study, we examined the mechanism of action of monoterpenes perillyl alcohol (POH) and perillic acid (PA) upon in vitro replication of HSV-1 KOS wild-type and the syn-mutant 17+ strain on Vero cells by plaque assay. RESULTS The cytotoxicity of POH and PA was measured by MTT assay and indicated that both compounds had high anti-HSV-1 activities in a concentration range that was not toxic for Vero cells. In addition, PCR analysis showed that POH and PA did not inhibit viral genome replication, but rather the release of infective virion particles from Vero cells. CONCLUSIONS Such findings suggest that POH and PA exert action upon late stages of HSV-1 maturation, therefore, indicating a promising perspective to its application in clinical investigation as effective anti-HSV-1 therapy preventing intermittent reactivation and progressive grey matter loss.
Collapse
Affiliation(s)
- Camilly Pires Mello
- Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, RJ, Brazil.,Department of Microbiology and Molecular Genetics, College of Medicine, University of Florida, Gainesville, FL, USA.,Present address: NanoScience Technology Center, University of Central Florida, Orlando, FL, USA
| | - Thereza Quirico-Santos
- Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Lídia Fonte Amorim
- Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Viveca Giongo Silva
- Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Lucianne Madeira Fragel
- Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, RJ, Brazil
| | - David C Bloom
- Department of Microbiology and Molecular Genetics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Izabel Palmer Paixão
- Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, RJ, Brazil.,Department of Microbiology and Molecular Genetics, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
14
|
Herbal Gel Formulation Developed for Anti-Human Immunodeficiency Virus (HIV)-1 Activity Also Inhibits In Vitro HSV-2 Infection. Viruses 2018; 10:v10110580. [PMID: 30352961 PMCID: PMC6266149 DOI: 10.3390/v10110580] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/09/2018] [Accepted: 10/12/2018] [Indexed: 11/25/2022] Open
Abstract
Herpes simplex virus-2 (HSV-2) infection is the most common cause of genital ulcers. The impact of ulcers also demonstrates a strong link to the human immunodeficiency virus (HIV) infection. Complications, drug resistance, and side-effects of anti-viral drugs make the treatment of HSV-2 infection challenging. Herbal medicines have shown potential against HSV-2 and HIV infections. In this context, polyherbal gel formulation comprising 50% ethanolic extracts from Acacia catechu, Lagerstroemia speciosa, Terminalia chebula and Phyllanthus emblica has been developed. The gel formulation significantly exhibited virucidal activity against both HIV-1 and HSV-2 infections with IC50, 55.93 ± 5.30 µg/mL and 27.26 ± 4.87 µg/mL, respectively. It also inhibited HSV-2 attachment and penetration to the Vero cells with an IC50 = 46.55 ± 1.25 µg/mL and 54.94 ± 2.52 µg/mL respectively, which were significantly lower than acyclovir. However, acyclovir is more potent in post-infection assay with an IC50 = 0.065 ± 0.01 µg/mL whereas gel formulation showed an IC50 = 469.05 ± 16.65 µg/mL under similar conditions. Gel formulation showed no inhibitory effect on the viability of lactobacilli, human vaginal keratinocyte cells (Vk2/E6E7), and the integrity of the Caco-2 cells monolayer. Gel formulation did not lead to any significant increase in the secretion of pro-inflammatory cytokines and mutagenic index. The proposed gel formulation may be a promising candidate microbicide for the prevention of sexually transmitted HIV-1 and HSV-2.
Collapse
|
15
|
Young S, Body B, Moore F, Dunbar S. Multicenter evaluation of the Luminex® ARIES® HSV 1&2 Assay for the detection of herpes simplex virus types 1 and 2 in cutaneous and mucocutaneous lesion specimens. Expert Rev Mol Diagn 2016; 16:1241-1249. [PMID: 27771977 DOI: 10.1080/14737159.2016.1251841] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The ARIES® HSV 1&2 Assay is a new FDA cleared real-time PCR test for detection and differentiation of HSV-1 and HSV-2 DNA from cutaneous and mucocutaneous lesions. The test is performed on the ARIES® System, an automated sample to answer real-time PCR instrument that provides a closed system and simple workflow for performing molecular testing. Areas covered: This article reports the clinical performance of the ARIES® HSV 1&2 Assay assessed on 1963 prospectively collected specimens. Assay sensitivities were 91.1-95% (cutaneous) and 97-98.5% (mucocutaneous), and specificities were 88.8-94.2% (cutaneous) and 93.2-95.4% (mucocutaneous), as compared to the ELVIS® HSV test system. Expert commentary: Detection of HSV DNA by PCR is rapid and more sensitive than traditional culture and immunoassay methods and is being widely adopted in many laboratory settings. Sample to answer molecular platforms like ARIES® will enable routine and non-molecular labs to perform sensitive and rapid molecular testing with ease.
Collapse
Affiliation(s)
- Stephen Young
- a TriCore Reference Laboratories , Research and Clinical Trials , Albuquerque , NM , USA
| | - Barbara Body
- b Laboratory Corporation of America , Atlantic Division Regional Laboratory & Center for Esoteric Testing , Burlington , NC , USA
| | - Franklin Moore
- c Baystate Health , Molecular and Microbiology Laboratories , Springfield , MA , USA
| | - Sherry Dunbar
- d Luminex Corporation, Global Scientific Affairs , Austin , TX , USA
| |
Collapse
|
16
|
ATPase activity measurement of DNA replicative helicase from Bacillus stearothermophilus by malachite green method. Anal Biochem 2016; 509:46-49. [PMID: 27372608 DOI: 10.1016/j.ab.2016.06.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 11/24/2022]
Abstract
The DnaB helicase from Bacillus stearothermophilus (DnaBBst) was a model protein for studying the bacterial DNA replication. In this work, a non-radioactive method for measuring ATPase activity of DnaBBst helicase was described. The working parameters and conditions were optimized. Furthermore, this method was applied to investigate effects of DnaG primase, ssDNA and helicase loader protein (DnaI) on ATPase activity of DnaBBst. Our results showed this method was sensitive and efficient. Moreover, it is suitable for the investigation of functional interaction between DnaB and related factors.
Collapse
|
17
|
Sanchez MD, Ochoa AC, Foster TP. Development and evaluation of a host-targeted antiviral that abrogates herpes simplex virus replication through modulation of arginine-associated metabolic pathways. Antiviral Res 2016; 132:13-25. [PMID: 27192555 DOI: 10.1016/j.antiviral.2016.05.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 04/21/2016] [Accepted: 05/13/2016] [Indexed: 11/16/2022]
Abstract
Since their inception five decades ago, most antivirals have been engineered to disrupt a single viral protein or process that is essential for viral replication. This approach has limited the overall therapeutic effectiveness and applicability of current antivirals due to restricted viral specificity, a propensity for development of drug resistance, and an inability to control deleterious host-mediated inflammation. As obligate intracellular parasites, viruses are reliant on host metabolism and macromolecular synthesis pathways. Of these biosynthetic processes, many viruses, including Herpes simplex viruses (HSV), are absolutely dependent on the bioavailability of arginine, a non-essential amino acid that is critical for many physiological and pathophysiological processes associated with either facilitating viral replication or progression of disease. To assess if targeting host arginine-associated metabolic pathways would inhibit HSV replication, a pegylated recombinant human Arginase I (peg-ArgI) was generated and its in vitro anti-herpetic activity was evaluated. Cells continuously treated with peg-ArgI for over 48 h exhibited no signs of cytotoxicity or loss of cell viability. The antiviral activity of peg-ArgI displayed a classical dose-response curve with IC50's in the sub-nanomolar range. peg-ArgI potently inhibited HSV-1 and HSV-2 viral replication, infectious virus production, cell-to-cell spread/transmission and virus-mediated cytopathic effects. Not unexpectedly given its host-targeted mechanism of action, peg-ArgI showed similar effectiveness at controlling replication of single and multidrug resistant HSV-1 mutants. These findings illustrate that targeting host arginine-associated metabolic pathways is an effective means of controlling viral replicative processes. Further exploration into the breadth of viruses inhibited by peg-ArgI, as well as the ability of peg-ArgI to suppress arginine-associated virus-mediated pathophysiological disease processes is warranted.
Collapse
Affiliation(s)
- Maria Dulfary Sanchez
- Department of Microbiology, Immunology, and Parasitology, School of Medicine, Louisiana State University Health Sciences Center, USA; Department of Pediatrics, School of Medicine, Louisiana State University Health Sciences Center, USA; The Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, USA
| | - Augusto C Ochoa
- Department of Pediatrics, School of Medicine, Louisiana State University Health Sciences Center, USA; The Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, USA; The Louisiana Vaccine Center, New Orleans, LA, 70112, USA
| | - Timothy P Foster
- Department of Microbiology, Immunology, and Parasitology, School of Medicine, Louisiana State University Health Sciences Center, USA; Department of Ophthalmology, School of Medicine, Louisiana State University Health Sciences Center, USA; The Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, USA; The Louisiana Vaccine Center, New Orleans, LA, 70112, USA.
| |
Collapse
|
18
|
Abstract
Herpes simplex virus (HSV), a member of the Herpesviridae family, is a significant human pathogen that results in mucocutaneous lesions in the oral cavity or genital infections. Acyclovir (ACV) and related nucleoside analogues can successfully treat HSV infections, but the emergence of drug resistance to ACV has created a barrier for the treatment of HSV infections, especially in immunocompromised patients. There is an urgent need to explore new and effective tactics to circumvent drug resistance to HSV. This review summarises the current strategies in the development of new targets (the DNA helicase/primase (H/P) complex), new types of molecules (nature products) and new antiviral mechanisms (lethal mutagenesis of Janus-type nucleosides) to fight the drug resistance of HSV.
Collapse
|
19
|
Piccirillo A, Lavezzo E, Niero G, Moreno A, Massi P, Franchin E, Toppo S, Salata C, Palù G. Full Genome Sequence-Based Comparative Study of Wild-Type and Vaccine Strains of Infectious Laryngotracheitis Virus from Italy. PLoS One 2016; 11:e0149529. [PMID: 26890525 PMCID: PMC4758665 DOI: 10.1371/journal.pone.0149529] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/02/2016] [Indexed: 02/07/2023] Open
Abstract
Infectious laryngotracheitis (ILT) is an acute and highly contagious respiratory disease of chickens caused by an alphaherpesvirus, infectious laryngotracheitis virus (ILTV). Recently, full genome sequences of wild-type and vaccine strains have been determined worldwide, but none was from Europe. The aim of this study was to determine and analyse the complete genome sequences of five ILTV strains. Sequences were also compared to reveal the similarity of strains across time and to discriminate between wild-type and vaccine strains. Genomes of three ILTV field isolates from outbreaks occurred in Italy in 1980, 2007 and 2011, and two commercial chicken embryo origin (CEO) vaccines were sequenced using the 454 Life Sciences technology. The comparison with the Serva genome showed that 35 open reading frames (ORFs) differed across the five genomes. Overall, 54 single nucleotide polymorphisms (SNPs) and 27 amino acid differences in 19 ORFs and two insertions in the UL52 and ORFC genes were identified. Similarity among the field strains and between the field and the vaccine strains ranged from 99.96% to 99.99%. Phylogenetic analysis revealed a close relationship among them, as well. This study generated data on genomic variation among Italian ILTV strains revealing that, even though the genetic variability of the genome is well conserved across time and between wild-type and vaccine strains, some mutations may help in differentiating among them and may be involved in ILTV virulence/attenuation. The results of this study can contribute to the understanding of the molecular bases of ILTV pathogenicity and provide genetic markers to differentiate between wild-type and vaccine strains.
Collapse
Affiliation(s)
- Alessandra Piccirillo
- Department of Comparative Biomedicine and Food Science (BCA), University of Padua, Legnaro (Padua), Italy
- * E-mail:
| | - Enrico Lavezzo
- Department of Molecular Medicine, University of Padua (DMM), Padua, Italy
| | - Giulia Niero
- Department of Comparative Biomedicine and Food Science (BCA), University of Padua, Legnaro (Padua), Italy
| | - Ana Moreno
- Department of Virology, Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna (IZSLER), Brescia, Italy
| | - Paola Massi
- Department of Diagnostics, Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna (IZSLER), Forlì, Italy
| | - Elisa Franchin
- Department of Molecular Medicine, University of Padua (DMM), Padua, Italy
| | - Stefano Toppo
- Department of Molecular Medicine, University of Padua (DMM), Padua, Italy
| | - Cristiano Salata
- Department of Molecular Medicine, University of Padua (DMM), Padua, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padua (DMM), Padua, Italy
| |
Collapse
|
20
|
Vashishtha AK, Kuchta RD. Effects of Acyclovir, Foscarnet, and Ribonucleotides on Herpes Simplex Virus-1 DNA Polymerase: Mechanistic Insights and a Novel Mechanism for Preventing Stable Incorporation of Ribonucleotides into DNA. Biochemistry 2016; 55:1168-77. [PMID: 26836009 DOI: 10.1021/acs.biochem.6b00065] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We examined the impact of two clinically approved anti-herpes drugs, acyclovir and Forscarnet (phosphonoformate), on the exonuclease activity of the herpes simplex virus-1 DNA polymerase, UL30. Acyclovir triphosphate and Foscarnet, along with the closely related phosphonoacetic acid, did not affect exonuclease activity on single-stranded DNA. Furthermore, blocking the polymerase active site due to either binding of Foscarnet or phosphonoacetic acid to the E-DNA complex or polymerization of acyclovir onto the DNA also had a minimal effect on exonuclease activity. The inability of the exonuclease to excise acyclovir from the primer 3'-terminus results from the altered sugar structure directly impeding phosphodiester bond hydrolysis as opposed to inhibiting binding, unwinding of the DNA by the exonuclease, or transfer of the DNA from the polymerase to the exonuclease. Removing the 3'-hydroxyl or the 2'-carbon from the nucleotide at the 3'-terminus of the primer strongly inhibited exonuclease activity, although addition of a 2'-hydroxyl did not affect exonuclease activity. The biological consequences of these results are twofold. First, the ability of acyclovir and Foscarnet to block dNTP polymerization without impacting exonuclease activity raises the possibility that their effects on herpes replication may involve both direct inhibition of dNTP polymerization and exonuclease-mediated destruction of herpes DNA. Second, the ability of the exonuclease to rapidly remove a ribonucleotide at the primer 3'-terminus in combination with the polymerase not efficiently adding dNTPs onto this primer provides a novel mechanism by which the herpes replication machinery can prevent incorporation of ribonucleotides into newly synthesized DNA.
Collapse
Affiliation(s)
- Ashwani Kumar Vashishtha
- Department of Chemistry and Biochemistry, University of Colorado , Boulder, Colorado 80309-0215, United States
| | - Robert D Kuchta
- Department of Chemistry and Biochemistry, University of Colorado , Boulder, Colorado 80309-0215, United States
| |
Collapse
|
21
|
Collot M, Rouard C, Brunet C, Agut H, Boutolleau D, Burrel S. High conservation of herpes simplex virus UL5/UL52 helicase-primase complex in the era of new antiviral therapies. Antiviral Res 2016; 128:1-6. [PMID: 26826441 DOI: 10.1016/j.antiviral.2016.01.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/13/2016] [Accepted: 01/22/2016] [Indexed: 10/22/2022]
Abstract
The emergence of herpes simplex virus (HSV) resistance to current antiviral drugs, that all target the viral DNA polymerase, constitutes a major obstacle to antiviral treatment effectiveness of HSV infections, especially in immunocompromised patients. A novel and promising class of inhibitors of the HSV UL5/UL52 helicase-primase (HP) complex has been reported to hinder viral replication with a high potency. In this study, we describe the low natural polymorphism (interstrain identity >99.1% at both nucleotide and amino acid levels) of HSV HP complex subunits pUL5 and pUL52 among 64 HSV (32 HSV-1 and 32 HSV-2) clinical isolates, and we show that the HSV resistance profile to the first-line antiviral drug acyclovir (ACV) does not impact on the natural polymorphism of HSV HP complex. Genotypic tools and polymorphism data concerning HSV HP complex provided herein will be useful to detect drug resistance mutations in a relevant time frame when HP inhibitors (HPIs), i.e., amenamevir and pritelivir, will be available in medical practice.
Collapse
Affiliation(s)
- Marianne Collot
- Centre Hospitalier et Universitaire de Nantes, Service de Virologie, Nantes, France; EA4271 « Immunovirology and Genetic Polymorphism », Université de Nantes, France
| | - Caroline Rouard
- AP-HP, Hôpital Universitaire Antoine Béclère, Service de Microbiologie, Clamart, France; EA4043 - Unité Bactéries Pathogènes et Santé UFR de Pharmacie-Université Paris-Sud, France
| | - Christel Brunet
- AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, Service de Virologie, Paris, France
| | - Henri Agut
- AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, Service de Virologie, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), INSERM U1135, Paris, France
| | - David Boutolleau
- AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, Service de Virologie, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), INSERM U1135, Paris, France
| | - Sonia Burrel
- AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, Service de Virologie, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), INSERM U1135, Paris, France.
| |
Collapse
|
22
|
Fernández-Menéndez S, Fernández-Morán M, Fernández-Vega I, Pérez-Álvarez A, Villafani-Echazú J. Epstein-Barr virus and multiple sclerosis. From evidence to therapeutic strategies. J Neurol Sci 2016; 361:213-9. [PMID: 26810546 DOI: 10.1016/j.jns.2016.01.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/21/2015] [Accepted: 01/05/2016] [Indexed: 10/22/2022]
Abstract
Multiple sclerosis is caused by a complex interaction between genetic predisposition and environmental factors. Epstein-Barr virus (EBV) is an environmental risk factor that is strongly related to multiple sclerosis (MS), since EBV seropositivity is linked to a significant risk of developing MS. EBV may be involved in the pathogenesis of the disease and it is possibly a prerequisite for the development of MS. EBV infection persists in B-cells during the lifetime of the host and can modulate their function. In addition, MS patients might have a deficient capacity to eliminate latent EBV infection in the central nervous system and this would promote the accumulation of infected B cells. Several mechanisms of pathogenesis, including a direct and indirect function of infected B cells, have been postulated in inflammation and neurodegeneration. A relationship between EBV and human endogenous retroviruses in the pathogenesis of MS has also been reported. If EBV is important in the pathogenesis of MS, different therapeutic strategies seem possible for MS treatment.
Collapse
Affiliation(s)
| | - Marta Fernández-Morán
- Department of Neuropaediatrics, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Iván Fernández-Vega
- Pathology department (Neuropathology division), Hospital Universitario Araba, Álava, Spain
| | - Angel Pérez-Álvarez
- Department of Neurology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | |
Collapse
|
23
|
López-Labrador FX, Berenguer M, Navarro D. Overcoming drug resistance in HSV, CMV, HBV and HCV infection. Future Microbiol 2015; 10:1759-66. [PMID: 26522939 DOI: 10.2217/fmb.15.74] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although vaccination has provided as a very efficient preventive tool, antiviral therapy is still needed to control viral infections not avoidable by prophylaxis with vaccines; those caused by viruses for which a vaccine is available, but vaccination is not universally implemented or does not result in complete, long-term protection; and in immunocompromised individuals with reduced immune control of viral replication. After more than 50 years of the first licensing for an antiherpetic drug, novel compounds for herpes-simplex viruses and human cytomegalovirus will open new strategies for better control and management of these two recurrent viral infections. Besides, the development and use of antiviral drugs for hepatitis viruses causing chronic liver disease has evolved from the unavailability of compounds targeting virus-specific pathways, to the generalization of different treatment regimes based on specific antiviral drugs, both for hepatitis B and C viruses. However, due to the nature of the viral genomes and their replication, in the face of the selective pressure imposed by antiviral drugs viral-resistant variants inevitably emerge, and specific strategies are needed to avoid, or reduce, their clinical and public health impact.
Collapse
Affiliation(s)
- F Xavier López-Labrador
- Virology Laboratory, and Joint Units on Evolution and Health & Genomics and Health, FISABIO-Public Health, Generalitat Valenciana/Institut Cavanilles, University of Valencia, Spain.,CIBER-ESP, Instituto de Salud Carlos III, Spain
| | - Marina Berenguer
- Digestive Diseases Service (Hepatology), Hospital Universitari i Politècnic La Fe de Valencia; and Department of Medicine, Medical School, University of Valencia, Spain.,CIBER-EHD, Instituto de Salud Carlos III, Spain
| | - David Navarro
- Microbiology Service, Hospital Clínic Universitari de Valencia, Spain.,Department of Microbiology & Ecology, University of Valencia, Spain
| |
Collapse
|
24
|
Regulated bioanalysis of conformers - A case study with ASP2151 in dog plasma and urine. J Chromatogr B Analyt Technol Biomed Life Sci 2015; 997:56-63. [PMID: 26093120 DOI: 10.1016/j.jchromb.2015.05.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 05/22/2015] [Accepted: 05/25/2015] [Indexed: 11/23/2022]
Abstract
We developed and validated bioanalytical methods for a potent helicase-primase inhibitor ASP2151 that has two conformers. These conformers elute as unseparated broad peaks under ordinary high-performance liquid chromatographic conditions, indicating discernable differences in hydrophobicity. We observed that column temperature and mobile phase pH have no effect on these peaks and that conformers form a single symmetrical peak when tetrahydrofuran is added to the mobile phase. In addition, we needed to develop semi-automated methods where inter-conversion of the conformers is unlikely to cause sample-to-sample extraction variability. Briefly, following the addition of deuterium-labeled ASP2151 as an internal standard (IS), dog plasma samples or acetonitrile-added urine samples were filtrated. The filtrates were then injected into a column-switching liquid chromatography-tandem mass spectrometry (LC-MS/MS) system and trapped onto an extraction column. Extracts were back-flushed onto an analytical C18 column (4.6×50mm, 3μm) with a mobile phase consisting of methanol, tetrahydrofuran, and 20mmol/L ammonium acetate (45:5:50, v/v/v). The eluent was monitored in the negative atmospheric pressure chemical ionization mode. The calibration curve was linear over a range of 5-1000ng/mL for plasma and 0.5-100μg/mL for urine. Validation data met the acceptance criteria in accordance with regulatory guidance and demonstrated that these methods were selective, accurate, and reproducible. In addition, the present methods were successfully applied to a pharmacokinetic study in dogs.
Collapse
|
25
|
Ahmad M, Tarique M, Afrin F, Tuteja N, Tuteja R. Identification of inhibitors of Plasmodium falciparum RuvB1 helicase using biochemical assays. PROTOPLASMA 2015; 252:117-125. [PMID: 24934654 DOI: 10.1007/s00709-014-0664-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 06/06/2014] [Indexed: 06/03/2023]
Abstract
Human malaria is a major parasitic infection, and the situation has worsened mainly due to the emergence of resistant malaria parasites to several anti-malarial drugs. Thus, an urgent need to find suitable drug targets has led to the development of newer classes of anti-malarial drugs. Helicases have been targeted to develop therapeutics for viral, bacterial, and other microorganism infections. Recently, Plasmodium falciparum RuvB ATPases/helicases have been characterized and proposed as a suitable antimalarial drug target. In the present study, the screening of various compounds was done and the results suggest that PfRuvB1 ATPase activity is inhibited considerably by the novobiocin and partially by cisplatin and ciprofloxacin. Helicase assay of PfRuvB1 in the presence of various compounds suggest novobiocin, actinomycin, and ethidium bromide as potent inhibitors. Novobiocin inhibits the helicase activity of PfRuvB1 possibly by blocking the ATPase activity of PfRuvB1. This study is unique in respect to the identification of novobiocin as inhibitor of PfRuvB1, partially by competing with ATP binding at its active site and provides evidence for PfRuvB1 as target of novobiocin after DNA gyrase-B and HSP90. These studies will certainly help the pharmacologist to design and develop some novel inhibitor specific to PfRuvB1, which may serve as suitable chemotherapeutics to target malaria.
Collapse
Affiliation(s)
- Moaz Ahmad
- Malaria Group, International Centre for Genetic Engineering and Biotechnology, P.O. Box 10504, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | | | | | | | | |
Collapse
|
26
|
Muylaert I, Zhao Z, Elias P. UL52 primase interactions in the herpes simplex virus 1 helicase-primase are affected by antiviral compounds and mutations causing drug resistance. J Biol Chem 2014; 289:32583-92. [PMID: 25278021 PMCID: PMC4239612 DOI: 10.1074/jbc.m114.609453] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 09/29/2014] [Indexed: 12/25/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) UL5/8/52 helicase-primase complex is required for DNA unwinding at the replication fork and synthesis of primers during virus replication, and it has become a promising novel target for antiviral therapy. Using molecular cloning, we have identified three separate domains of UL52. Co-immunoprecipitation experiments in extracts from cells transiently expressing HA-tagged UL5, FLAG-UL8, and enhanced GFP-tagged UL52 domains revealed that the N-terminal domain of UL52 primase binds UL5 helicase and the middle domain interacts with the UL8 accessory protein. In addition, an interaction between the single strand DNA-binding protein ICP8 and the UL52 middle domain was observed. The complex between UL5 and UL52 was stabilized by the antiviral compound BAY 54-6322, and mutations providing resistance to the drug obliterate this effect. Our results also suggest a mechanism for accommodating conformational strain resulting from movement of UL5 and UL52 in opposite directions on the lagging strand template, and they identify molecular complexes that can be further examined by structural biology techniques to resolve the mechanism of primer synthesis during herpesvirus replication. Finally, they help to explain the mechanism of action of a novel class of antiviral compounds currently being evaluated in clinical trials.
Collapse
Affiliation(s)
- Isabella Muylaert
- From the Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Gothenburg, Box 440, SE-405 30 Gothenburg, Sweden
| | - Zhiyuan Zhao
- From the Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Gothenburg, Box 440, SE-405 30 Gothenburg, Sweden
| | - Per Elias
- From the Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Gothenburg, Box 440, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
27
|
James SH, Larson KB, Acosta EP, Prichard MN. Helicase-primase as a target of new therapies for herpes simplex virus infections. Clin Pharmacol Ther 2014; 97:66-78. [PMID: 25670384 DOI: 10.1002/cpt.3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 09/16/2014] [Indexed: 01/13/2023]
Abstract
The seminal discovery of acyclovir 40 years ago heralded the modern era of truly selective antiviral therapies and this drug remains the therapy of choice for herpes simplex virus infections. Yet by modern standards, its antiviral activity is modest and new drugs against novel molecular targets such as the helicase-primase have the potential to improve clinical outcome, particularly in high-risk patients. A brief synopsis of current therapies for these infections and clinical need is provided to help provide an initial perspective. The function of the helicase-primase complex is then summarized and the development of new inhibitors of the helicase-primase complex, such as pritelivir and amenamevir, is discussed. We review their mechanism of action, propensity for drug resistance, and pharmacokinetic characteristics and discuss their potential to advance current therapeutic options. Strategies that include combinations of these inhibitors with acyclovir are also considered, as they will likely maximize clinical efficacy.
Collapse
Affiliation(s)
- S H James
- Division of Infectious Diseases, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | | |
Collapse
|
28
|
Yoneda JD, Albuquerque MG, Leal KZ, Santos FDC, Batalha PN, Brozeguini L, Seidl PR, de Alencastro RB, Cunha AC, de Souza MCB, Ferreira VF, Giongo VA, Cirne-Santos C, Paixão IC. Docking of anti-HIV-1 oxoquinoline-acylhydrazone derivatives as potential HSV-1 DNA polymerase inhibitors. J Mol Struct 2014. [DOI: 10.1016/j.molstruc.2014.05.081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
29
|
Plasmodium falciparum UvrD activities are downregulated by DNA-interacting compounds and its dsRNA inhibits malaria parasite growth. BMC BIOCHEMISTRY 2014; 15:9. [PMID: 24707807 PMCID: PMC4234510 DOI: 10.1186/1471-2091-15-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/31/2014] [Indexed: 12/11/2022]
Abstract
Background Human malaria parasite infection and its control is a global challenge which is responsible for ~0.65 million deaths every year globally. The emergence of drug resistant malaria parasite is another challenge to fight with malaria. Enormous efforts are being made to identify suitable drug targets in order to develop newer classes of drug. Helicases play crucial roles in DNA metabolism and have been proposed as therapeutic targets for cancer therapy as well as viral and parasitic infections. Genome wide analysis revealed that Plasmodium falciparum possesses UvrD helicase, which is absent in the human host. Results Recently the biochemical characterization of P. falciparum UvrD helicase revealed that N-terminal UvrD (PfUDN) hydrolyses ATP, translocates in 3’ to 5’ direction and interacts with MLH to modulate each other’s activity. In this follow up study, further characterization of P. falciparum UvrD helicase is presented. Here, we screened the effect of various DNA interacting compounds on the ATPase and helicase activity of PfUDN. This study resulted into the identification of daunorubicin (daunomycin), netropsin, nogalamycin, and ethidium bromide as the potential inhibitor molecules for the biochemical activities of PfUDN with IC50 values ranging from ~3.0 to ~5.0 μM. Interestingly etoposide did not inhibit the ATPase activity but considerable inhibition of unwinding activity was observed at 20 μM. Further study for analyzing the importance of PfUvrD enzyme in parasite growth revealed that PfUvrD is crucial/important for its growth ex-vivo. Conclusions As PfUvrD is absent in human hence on the basis of this study we propose PfUvrD as suitable drug target to control malaria. Some of the PfUvrD inhibitors identified in the present study can be utilized to further design novel and specific inhibitor molecules.
Collapse
|
30
|
Affiliation(s)
- Richard J Whitley
- From the Departments of Pediatrics, Microbiology, Medicine, and Neurosurgery, University of Alabama at Birmingham
| | | |
Collapse
|
31
|
Paintsil E, Cheng YC. Antiviral Agents☆. REFERENCE MODULE IN BIOMEDICAL SCIENCES 2014. [PMCID: PMC7150273 DOI: 10.1016/b978-0-12-801238-3.02387-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Antiviral agents are drugs approved in the USA by the Food and Drug Administration (FDA) for the treatment or control of viral infections. Available antiviral agents mainly target stages in the viral life cycle. The target stages in the viral life cycle are; viral attachment to host cell, uncoating, synthesis of viral mRNA, translation of mRNA, replication of viral RNA and DNA, maturation of new viral proteins, budding, release of newly synthesized virus, and free virus in body fluids. Two important factors that can limit the utility of antiviral drugs are toxicity and the development of resistance to the antiviral agent by the virus. In addition, host phenotypic behaviors toward antiviral drugs because of either genomic or epigenetic factors could limit the efficacy of an antiviral agent in an individual. This article summarizes the most relevant pharmacologic and clinical properties of current antiviral agents, and targets for novel antiviral agents.
Collapse
|
32
|
Field HJ. A short commentary on the review by Weller and Kuchta: the DNA helicase-primase complex as a target for herpes virus infection. Expert Opin Ther Targets 2013; 17:1113-4. [PMID: 24044544 DOI: 10.1517/14728222.2013.840294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|