1
|
Dahl DK, Srinivasan P, Janusziewicz R, King JL, Shrivastava R, Zhang J, Little D, Bachman S, Kelley K, Cottrell ML, Schauer AP, Sykes C, Kashuba ADM, Smith J, Benhabbour SR. Next-generation 3D printed multipurpose prevention intravaginal ring for prevention of HIV, HSV-2, and unintended pregnancy. J Control Release 2024; 376:1209-1224. [PMID: 39500407 DOI: 10.1016/j.jconrel.2024.10.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/14/2024]
Abstract
Globally, nearly half of all pregnancies are unintended, ∼1.3 million new human immunodeficiency virus (HIV) infections are reported every year, and more than 500 million people are estimated to have a genital herpes simplex virus (HSV-2) infection. Here we report the first 3D printed multipurpose prevention technology (MPT) intravaginal ring (IVR) for prevention of HIV, HSV-2, and unintended pregnancy. The IVRs were fabricated using state-of-the-art Continuous Liquid Interface Production (CLIP™) 3D printing technology using a biocompatible silicone-urethane based resin. Anti-HIV drug (Dapivirine, DPV), anti-herpes drug (Pritelivir, PTV) and a contraceptive drug (Levonorgestrel, LNG) were loaded in a macaque size IVR (25 mm outer diameter, OD; 6.0 mm cross-section, CS) allometrically scaled from the human size (54 mm OD; 7.6 mm CS) IVR analogue. All three active pharmaceutical ingredients (APIs) were loaded in the IVR using a single-step drug loading process driven by absorption. DPV, PTV, and LNG elicited zero-order release kinetics in vitro in simulated vaginal fluid (SVF) at pH 4 and pH 8 relevant to human and macaque vaginal pH respectively. CLIP 3D printed MPT IVRs remained stable after 6 months of storage at 4 °C with no change in physical, dimensional, or mechanical properties and no change in drug concentration and absence of drug degradation byproducts. The MPT IVRs elicited sustained release of all three APIs in macaques for 28 days with median plasma concentrations of 138 pg/mL (DPV), 18,700 pg/mL (PTV), and 335 pg/mL (LNG). Safety studies demonstrated that the MPT IVRs were safe and well tolerated in the macaques with no observed change or abnormalities in vaginal pH and no significant changes in any of the 22 mucosal cytokines and chemokines tested including pro-inflammatory (IL-1β, IL-6, IL-8, IFN-γ, TNF-α, IL-17, IL-18) and anti-inflammatory (IL-10, IL-12) cytokines while the MPT IVR was in place or after its removal. Additionally, MPT IVRs elicited no observed alterations in systemic CD4+ and CD8+ T cells during the entire study. Collectively, the proposed MPT IVR has potential to expand preventative choices for young women and girls against unintended pregnancy and two highly prevalent sexually transmitted infections (STIs).
Collapse
Affiliation(s)
- Denali K Dahl
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Priya Srinivasan
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Rima Janusziewicz
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jasmine L King
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Roopali Shrivastava
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jining Zhang
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Dawn Little
- Katmai Government Services (KGS), Anchorage, AK, USA
| | | | | | - Mackenzie L Cottrell
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amanda P Schauer
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Craig Sykes
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Angela D M Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - James Smith
- Laboratory Branch, Division of HIV Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - S Rahima Benhabbour
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
2
|
Erb-Zohar K, Bonsmann S, Pausch J, Sumner M, Birkmann A, Zimmermann H, Halabi A, Kropeit D. Effect of Renal and Hepatic Impairment on the Pharmacokinetics of Pritelivir and Its Metabolites. Clin Pharmacol Drug Dev 2024; 13:1198-1211. [PMID: 39318306 DOI: 10.1002/cpdd.1469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/13/2024] [Indexed: 09/26/2024]
Abstract
Two trials were performed to evaluate the effect of renal and hepatic impairment on the pharmacokinetics of pritelivir and its metabolites. The renal impairment trial included subjects with mild, moderate, and severe impairment, while the hepatic impairment trial included subjects with moderate impairment. Both trials recruited a matched control group of healthy subjects. Following a single oral dose of 100 mg of pritelivir, mild and moderate renal impairment and moderate hepatic impairment did not have a clinically relevant effect on the pharmacokinetics of pritelivir. In subjects with severe renal impairment, pritelivir exposure (area under the plasma concentration-time curve from time 0 to infinity (AUC0- inf) was 57% higher compared with controls. Pritelivir plasma protein binding was similar in subjects and controls with renal impairment, while the free fraction was higher in subjects with moderate hepatic impairment, increasing unbound pritelivir exposure by 23%. For the metabolites pyridinyl phenyl acetic acid (PPA), amino thiazole sulfonamide (ATS), and PPA-acyl glucuronide, generally higher exposure was observed with increasing degree of renal impairment (ie, moderate to severe), but not with mild impairment. A modest effect of moderate hepatic impairment was observed for PPA and ATS. Pritelivir was safe and well tolerated in healthy subjects and subjects with renal or hepatic impairment.
Collapse
Affiliation(s)
| | | | - Jörg Pausch
- Affiliation at time of the studies: Aicuris Anti-infective Cures AG, Wuppertal, Germany
| | | | | | | | - Atef Halabi
- CRS Clinical Research Services Kiel GmbH, Kiel, Germany
| | - Dirk Kropeit
- AiCuris Anti-infective Cures AG, Wuppertal, Germany
| |
Collapse
|
3
|
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
|
4
|
Sallée L, Boutolleau D. Management of Refractory/Resistant Herpes Simplex Virus Infections in Haematopoietic Stem Cell Transplantation Recipients: A Literature Review. Rev Med Virol 2024; 34:e2574. [PMID: 39090526 DOI: 10.1002/rmv.2574] [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: 02/07/2024] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Herpes simplex virus (HSV) infections in allogeneic haematopoietic stem cell transplantation (HSCT) recipients pose significant challenges, with higher incidence, severity, and risk of emergence of resistance to antivirals due to impaired T-cell mediated immunity. This literature review focuses on acyclovir-refractory/resistant HSV infections in HSCT recipients. The review addresses the efficacy of antiviral prophylaxis, the incidence of acyclovir-refractory/resistant HSV infections, and the identification of risk factors and potential prognostic impact associated with those infections. Additionally, alternative therapeutic options are discussed. While acyclovir prophylaxis demonstrates a significant benefit in reducing HSV infections in HSCT recipients and, in some cases, overall mortality, concerns arise about the emergence of drug-resistant HSV strains. Our systematic review reports a median incidence of acyclovir-resistant HSV infections of 16.1%, with an increasing trend in recent years. Despite limitations in available studies, potential risk factors of emergence of HSV resistance to acyclovir include human leucocyte antigen (HLA) mismatches, myeloid neoplasms and acute leukaemias, and graft-versus-host disease (GVHD). Limited evidences suggest a potentially poorer prognosis for allogeneic HSCT recipients with acyclovir-refractory/resistant HSV infection. Alternative therapeutic approaches, such as foscarnet, cidofovir, topical cidofovir, optimised acyclovir dosing, and helicase-primase inhibitors offer promising options but require further investigations. Overall, larger studies are needed to refine preventive and therapeutic strategies for acyclovir-refractory/resistant HSV infections in allogeneic HSCT recipients and to identify those at higher risk.
Collapse
Affiliation(s)
- Léo Sallée
- Assistance Publique-Hôpitaux de Paris, Université Sorbonne Paris Nord, Paris, France
| | - David Boutolleau
- AP-HP, Centre National de Référence Herpèsvirus (Laboratoire Associé), Hôpital Pitié-Salpêtrière, Service de Virologie, and Sorbonne Université, INSERM, UMR-S 1136, Institut Pierre Louis d'Épidémiologie et de Santé Publique (iPLESP), Paris, France
| |
Collapse
|
5
|
de Vries M, Bonsmann S, Pausch J, Sumner M, Birkmann A, Zimmermann H, Kropeit D. Evaluation of the Clinical Drug-Drug Interaction Potential of Pritelivir on Transporters and CYP450 Enzymes Using a Cocktail Approach. Clin Pharmacol Drug Dev 2024; 13:755-769. [PMID: 38752475 DOI: 10.1002/cpdd.1408] [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: 11/16/2023] [Accepted: 04/08/2024] [Indexed: 07/05/2024]
Abstract
Pritelivir is a novel viral helicase-primase inhibitor active against herpes simplex virus. In vitro drug-drug interaction studies indicated that pritelivir has the potential for clinically relevant interactions on the cytochrome P450 (CYP) enzymes 2C8, 2C9, 3A4, and 2B6, and intestinal uptake transporter organic anion transporting polypeptide (OATP) 2B1 and efflux transporter breast cancer resistance protein (BCRP). This was evaluated in 2 clinical trials. In 1 trial the substrates flurbiprofen (CYP2C9), bupropion (CYP2B6), and midazolam (CYP3A4) were administered simultaneously as part of the Geneva cocktail, while the substrate celiprolol (OAPT2B1) was administered separately. In another trial, the substrates repaglinide (CYP2C8) and rosuvastatin (BCRP) were administered separately. Exposure parameters of the substrates and their metabolites (flurbiprofen and bupropion only) were compared after administration with or without pritelivir under therapeutic concentrations. The results of these trials indicated that pritelivir has no clinically relevant effect on the exposure of substrates for the intestinal uptake transporter OATP2B1 and the CYP enzymes 3A4, 2B6, 2C9, and 2C8, and has a weak inhibitory effect on the intestinal efflux transporter BCRP. In summary, the results suggest that pritelivir has a low drug-drug interaction potential.
Collapse
Affiliation(s)
| | | | - Jörg Pausch
- AiCuris Anti-infective Cures AG, Wuppertal, Germany
- Present affiliation: BioNTech SE, Mainz, Germany
| | | | | | | | - Dirk Kropeit
- AiCuris Anti-infective Cures AG, Wuppertal, Germany
| |
Collapse
|
6
|
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
|
7
|
Connolly KL, Bachmann L, Hiltke T, Kersh EN, Newman LM, Wilson L, Mena L, Deal C. Summary of the Centers for Disease Control and Prevention/National Institute of Allergy and Infectious Diseases Joint Workshop on Genital Herpes: 3-4 November 2022. Open Forum Infect Dis 2024; 11:ofae230. [PMID: 38784760 PMCID: PMC11112275 DOI: 10.1093/ofid/ofae230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Genital herpes is caused by infection with herpes simplex virus types 1 and 2 (HSV-1 and HSV-2) and currently has no cure. The disease is the second-most common sexually transmitted infection in the United States, with an estimated 18.6 million prevalent genital infections caused by HSV-2 alone. Genital herpes diagnostics and treatments are not optimal, and no vaccine is currently available. The Centers for Disease Control and Prevention and the National Institute of Allergy and Infectious Diseases convened a workshop entitled "CDC/NIAID Joint Workshop on Genital Herpes." This report summarizes 8 sessions on the epidemiology of genital herpes, neonatal HSV, HSV diagnostics, vaccines, treatments, cures, prevention, and patient advocacy perspective intended to identify opportunities in herpes research and foster the development of strategies to diagnose, treat, cure, and prevent genital herpes.
Collapse
Affiliation(s)
- Kristie L Connolly
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Laura Bachmann
- Division of Sexually Transmitted Disease Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Thomas Hiltke
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Ellen N Kersh
- Division of Sexually Transmitted Disease Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Lori M Newman
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Lydia Wilson
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Leandro Mena
- Division of Sexually Transmitted Disease Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Carolyn Deal
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
8
|
Hammond SP, Rangaraju M, Sumner M, Timmler B, Chandrasekar P, Avery RK. A Multicenter Assessment of the Outcomes and Toxicities of Foscarnet for Treatment of Acyclovir-Resistant Mucocutaneous Herpes Simplex in Immunocompromised Patients. Open Forum Infect Dis 2024; 11:ofae046. [PMID: 38444818 PMCID: PMC10914364 DOI: 10.1093/ofid/ofae046] [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: 09/25/2023] [Accepted: 01/24/2024] [Indexed: 03/07/2024] Open
Abstract
Background Acyclovir-resistant mucocutaneous herpes simplex virus (HSV) infection is an uncommon problem typically seen in immunocompromised hosts. Systemic treatment options are limited. The performance of foscarnet and its toxicities in this population are poorly characterized. Methods This was a multicenter retrospective study of adults treated with foscarnet for HSV infection between January 2012 and December 2017. Relevant data were collected including demographics, baseline conditions, previous anti-HSV medications, concomitant medications, HSV outcomes, and adverse events. Acyclovir-resistant HSV infection was defined based on genotypic or phenotypic testing results; refractory infection was defined as infection not improving after 5 days of treatment-dosed antiviral therapy in those not tested for resistance. Results Twenty-nine patients had 31 episodes of HSV (15/18 resistant; among episodes without resistance testing, 7/10 refractory; 3 not evaluable) treated with foscarnet. All patients were immunocompromised including 19 (66%) with hematologic malignancy and 9 (31%) with HIV. Median duration of foscarnet was 16 days (range, 6-85 days). Fifteen episodes (48%) healed by the end of or after foscarnet. Median time to healing among those with resolution was 38 days (range, 9-1088 days). At least 1 adverse event during therapy was reported in 26 (84%) treatment episodes including 23 (74%) that were considered drug related. Common adverse events were electrolyte disturbance (20 [65%]) and kidney dysfunction (13 [42%]). Foscarnet was discontinued in 10 episodes (32%) due to an adverse event, including 6 due to kidney dysfunction. Conclusions Among 31 episodes of HSV treated with foscarnet, only half resolved with treatment, and adverse events were common.
Collapse
Affiliation(s)
- Sarah P Hammond
- Divisions of Infectious Diseases and Hematology/Oncology, Massachusetts General Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | - Pranatharthi Chandrasekar
- Karmanos Cancer Institute, Division of Infectious Diseases, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Robin K Avery
- Division of Infectious Diseases, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Chen LW, Wang SS, Chen LY, Huang HY, He SM, Hung CH, Lin CL, Chang PJ. Interaction and assembly of the DNA replication core proteins of Kaposi's sarcoma-associated herpesvirus. Microbiol Spectr 2023; 11:e0225423. [PMID: 37874136 PMCID: PMC10715029 DOI: 10.1128/spectrum.02254-23] [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: 05/30/2023] [Accepted: 09/21/2023] [Indexed: 10/25/2023] Open
Abstract
IMPORTANCE Eukaryotic DNA replication is a highly regulated process that requires multiple replication enzymes assembled onto DNA replication origins. Due to the complexity of the cell's DNA replication machinery, most of what we know about cellular DNA replication has come from the study of viral systems. Herein, we focus our study on the assembly of the Kaposi's sarcoma-associated herpesvirus core replication complex and propose a pairwise protein-protein interaction network of six highly conserved viral core replication proteins. A detailed understanding of the interaction and assembly of the viral core replication proteins may provide opportunities to develop new strategies against viral propagation.
Collapse
Affiliation(s)
- Lee-Wen Chen
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi, Taiwan
- Department of Pediatric Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Shie-Shan Wang
- Department of Pediatric Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Li-Yu Chen
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsiao-Yun Huang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Si-min He
- Department of Pediatric Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chien-Hui Hung
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Liang Lin
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Pey-Jium Chang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| |
Collapse
|
10
|
Bernstein DI, Sawtell NM, Bravo FJ, Dixon DA, Gege C, Kleymann G. Intermittent therapy with helicase-primase inhibitor IM-250 efficiently controls recurrent herpes disease and reduces reactivation of latent HSV. Antiviral Res 2023; 219:105733. [PMID: 37858763 DOI: 10.1016/j.antiviral.2023.105733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/21/2023]
Abstract
Herpes is a contagious life-long infection with persistently high incidence and prevalence, causing significant disease worldwide. Current therapies have efficacy against active HSV infections but no impact on the latent viral reservoir in neurons. Thus, despite treatment, disease recurs from latency and the infectious potential remains unaffected within patients. Here, efficacy of the helicase-primase inhibitor (HPI) IM-250 against chronic neuronal HSV infections utilizing two classic herpes in vivo latency/reactivation animal models (intravaginal guinea pig HSV-2 infection model and ocular mouse HSV-1 infection model) is presented. Intermittent therapy of infected animals with 4-7 cycles of IM-250 during latency silences subsequent recurrences analyzed up to 6 months. In contrast to common experience, our studies show that the latent reservoir is indeed accessible to antiviral therapy altering the latent viral reservoir such that reactivation frequency can be reduced significantly by prior IM-250 treatment. We provide evidence that antiviral treatment during HSV latency can reduce future reactivation from the latent reservoir, supporting a conceptual shift in the antiviral field, and reframing what is achievable with respect to therapy of latent neuronal HSV infections.
Collapse
Affiliation(s)
- David I Bernstein
- Cincinnati Children's Hospital Medical Center (CCHMC), University of Cincinnati, OH, USA
| | - Nancy M Sawtell
- Cincinnati Children's Hospital Medical Center (CCHMC), University of Cincinnati, OH, USA
| | - Fernando J Bravo
- Cincinnati Children's Hospital Medical Center (CCHMC), University of Cincinnati, OH, USA
| | - David A Dixon
- Cincinnati Children's Hospital Medical Center (CCHMC), University of Cincinnati, OH, USA
| | - Christian Gege
- Innovative Molecules GmbH, Lipowsky Str. 10, 81373, Munich, Bavaria, Germany
| | - Gerald Kleymann
- Innovative Molecules GmbH, Lipowsky Str. 10, 81373, Munich, Bavaria, Germany.
| |
Collapse
|
11
|
De Rose DU, Bompard S, Maddaloni C, Bersani I, Martini L, Santisi A, Longo D, Ronchetti MP, Dotta A, Auriti C. Neonatal herpes simplex virus infection: From the maternal infection to the child outcome. J Med Virol 2023; 95:e29024. [PMID: 37592873 DOI: 10.1002/jmv.29024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/03/2023] [Accepted: 07/27/2023] [Indexed: 08/19/2023]
Abstract
This review examines the recent literature on the management of herpes simplex virus (HSV) infections in neonates. We summarized the three clinical categories of maternal HSV infection during pregnancy (primary first episode, nonprimary first episode, or recurrent episode) and the mechanisms of fetal damage. Considering when the transmission of the infection from the mother to the fetus/newborn occurs, three types of neonatal infection can be distinguished: intrauterine infection (5% of cases), postnatal infection (10% of cases), and perinatal infections (85% of cases). Neonatal presentation could range from a limited disease with skin, eye, and mouth disease to central nervous system disease or disseminated disease: the treatment with acyclovir should be tailored according to symptoms and signs of infection, and virological tests. These children need a multidisciplinary follow-up, to timely intercept any deviation from normal neurodevelopmental milestones. Prevention strategies remain a challenge, in the absence of an available vaccine against HSV.
Collapse
Affiliation(s)
- Domenico Umberto De Rose
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, "Tor Vergata" University of Rome, Rome, Italy
| | - Sarah Bompard
- Neurorehabilitation Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Chiara Maddaloni
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Iliana Bersani
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Ludovica Martini
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Alessandra Santisi
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Daniela Longo
- Neuroradiology Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | | | - Andrea Dotta
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Cinzia Auriti
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
- Saint Camillus International University, Rome, Italy
| |
Collapse
|
12
|
Kropeit D, Bonsmann S, von Richter O, McCormick D, Pausch J, Sumner M, Birkmann A, Zimmermann H, Rübsamen-Schaeff H. First-in-Human, Single- and Multiple-Ascending-Dose, Food-Effect, and Absolute Bioavailability Trials to Assess the Pharmacokinetics, Safety, and Tolerability of Pritelivir, a Nonnucleoside Helicase-Primase Inhibitor Against Herpes Simplex Virus in Healthy Subjects. Clin Pharmacol Drug Dev 2023; 12:749-760. [PMID: 36860173 DOI: 10.1002/cpdd.1241] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/03/2023] [Indexed: 03/03/2023]
Abstract
The pharmacokinetics and safety of the novel herpes simplex virus helicase-primase inhibitor pritelivir were evaluated in 5 phase 1 trials: a single-ascending-dose trial, 2 multiple-ascending-dose trials, a food-effect trial, and an absolute bioavailability trial in healthy male subjects. One cohort of healthy female subjects was included in the single-ascending-dose trial. Pritelivir pharmacokinetics were linear up to 480 mg following single and up to 400 mg following multiple once-daily doses. The half-life ranged from 52 to 83 hours, and steady state was reached between 8 and 13 days. Maximum plasma concentration and area under the plasma concentration-time curve from time 0 to the last quantifiable concentration were 1.5- and 1.1-fold higher in female compared to male subjects. Absolute bioavailability was 72% under fasted conditions. Following a fatty diet, pritelivir time to maximum concentration was 1.5 hour delayed and maximum plasma concentration and area under the plasma concentration-time curve from time 0 to the last quantifiable concentration were 33% and 16% higher, respectively. Pritelivir was safe and well tolerated up to 600 mg following single and up to 200 mg following multiple once-daily doses. Considering a therapeutic dose of 100 mg once-daily, pritelivir demonstrated a favorable safety and tolerability and pharmacokinetic profile in healthy subjects to support further development.
Collapse
Affiliation(s)
- Dirk Kropeit
- Aicuris Anti-infective Cures AG, Wuppertal, Germany
| | | | - Oliver von Richter
- Global Clinical Development, Biopharmaceuticals, Hexal AG (A Sandoz Company), Holzkirchen, Germany
| | - David McCormick
- Aicuris Anti-infective Cures AG (Retired), Wuppertal, Germany
| | | | | | | | | | | |
Collapse
|
13
|
Srinivasan K, Ho DY. Case Commentary: "Salvage Treatment of Refractory HSV Oral Lesions with Pritelivir in Allogeneic Hematopoietic Cell Transplant Recipients" by Bosetti et al. Antimicrob Agents Chemother 2023; 67:e0027623. [PMID: 37014226 PMCID: PMC10190666 DOI: 10.1128/aac.00276-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Affiliation(s)
- Krithika Srinivasan
- Division of Infectious Diseases & Geographic Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Dora Y. Ho
- Division of Infectious Diseases & Geographic Medicine, School of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
14
|
Tian WJ, Wang XJ. Broad-Spectrum Antivirals Derived from Natural Products. Viruses 2023; 15:v15051100. [PMID: 37243186 DOI: 10.3390/v15051100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Scientific advances have led to the development and production of numerous vaccines and antiviral drugs, but viruses, including re-emerging and emerging viruses, such as SARS-CoV-2, remain a major threat to human health. Many antiviral agents are rarely used in clinical treatment, however, because of their inefficacy and resistance. The toxicity of natural products may be lower, and some natural products have multiple targets, which means less resistance. Therefore, natural products may be an effective means to solve virus infection in the future. New techniques and ideas are currently being developed for the design and screening of antiviral drugs thanks to recent revelations about virus replication mechanisms and the advancement of molecular docking technology. This review will summarize recently discovered antiviral drugs, mechanisms of action, and screening and design strategies for novel antiviral agents.
Collapse
Affiliation(s)
- Wen-Jun Tian
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, China Agricultural University, Beijing 100193, China
| | - Xiao-Jia Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, China Agricultural University, Beijing 100193, China
| |
Collapse
|
15
|
Sharma D, Sharma S, Akojwar N, Dondulkar A, Yenorkar N, Pandita D, Prasad SK, Dhobi M. An Insight into Current Treatment Strategies, Their Limitations, and Ongoing Developments in Vaccine Technologies against Herpes Simplex Infections. Vaccines (Basel) 2023; 11:vaccines11020206. [PMID: 36851084 PMCID: PMC9966607 DOI: 10.3390/vaccines11020206] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/05/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Herpes simplex virus (HSV) infection, the most prevalent viral infection that typically lasts for a lifetime, is associated with frequent outbreaks of oral and genital lesions. Oral herpes infection is mainly associated with HSV-1 through oral contact, while genital herpes originates due to HSV-2 and is categorized under sexually transmitted diseases. Immunocompromised patients and children are more prone to HSV infection. Over the years, various attempts have been made to find potential targets for the prevention of HSV infection. Despite the global distress caused by HSV infections, there are no licensed prophylactic and therapeutic vaccines available on the market against HSV. Nevertheless, there are numerous promising candidates in the pre-clinical and clinical stages of study. The present review gives an overview of two herpes viruses, their history, and life cycle, and different treatments adopted presently against HSV infections and their associated limitations. Majorly, the review covers the recent investigations being carried out globally regarding various vaccine strategies against oral and genital herpes virus infections, together with the recent and advanced nanotechnological approaches for vaccine development. Consequently, it gives an insight to researchers as well as people from the health sector about the challenges and upcoming solutions associated with treatment and vaccine development against HSV infections.
Collapse
Affiliation(s)
- Divya Sharma
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi 110017, India
| | - Supriya Sharma
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi 110017, India
| | - Natasha Akojwar
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India
| | - Ayusha Dondulkar
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India
| | - Nikhil Yenorkar
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India
| | - Deepti Pandita
- Delhi Institute of Pharmaceutical Sciences and Research, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi 110017, India
- Correspondence: (D.P.); (S.K.P.); (M.D.)
| | - Satyendra K. Prasad
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India
- Correspondence: (D.P.); (S.K.P.); (M.D.)
| | - Mahaveer Dhobi
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi 110017, India
- Correspondence: (D.P.); (S.K.P.); (M.D.)
| |
Collapse
|
16
|
Huntjens DW, Dijkstra JA, Verwiel LN, Slijkhuis M, Elbers P, Welkers MRA, Veldkamp AI, Kuijvenhoven MA, de Leeuw DC, Abdullah-Koolmees H, Kuipers MT, Bartelink IH. Optimizing Antiviral Dosing for HSV and CMV Treatment in Immunocompromised Patients. Pharmaceutics 2023; 15:pharmaceutics15010163. [PMID: 36678792 PMCID: PMC9863155 DOI: 10.3390/pharmaceutics15010163] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Herpes simplex virus (HSV) and cytomegalovirus (CMV) are DNA viruses that are common among humans. Severely immunocompromised patients are at increased risk of developing HSV or CMV disease due to a weakened immune system. Antiviral therapy can be challenging because these drugs have a narrow therapeutic window and show significant pharmacokinetic variability. Above that, immunocompromised patients have various comorbidities like impaired renal function and are exposed to polypharmacy. This scoping review discusses the current pharmacokinetic (PK) and pharmacodynamic (PD) knowledge of antiviral drugs for HSV and CMV treatment in immunocompromised patients. HSV and CMV treatment guidelines are discussed, and multiple treatment interventions are proposed: early detection of drug resistance; optimization of dose to target concentration by therapeutic drug monitoring (TDM) of nucleoside analogs; the introduction of new antiviral drugs; alternation between compounds with different toxicity profiles; and combinations of synergistic antiviral drugs. This research will also serve as guidance for future research, which should focus on prospective evaluation of the benefit of each of these interventions in randomized controlled trials.
Collapse
Affiliation(s)
- Daan W. Huntjens
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Jacob A. Dijkstra
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-20-444-3524
| | - Lisanne N. Verwiel
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Mirjam Slijkhuis
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Paul Elbers
- Department of Intensive Care Medicine, Laboratory for Critical Care Computational Intelligence (LCCI), Amsterdam Medical Data Science (AMDS), Amsterdam Cardiovascular Science (ACS), Amsterdam Institute for Infection and Immunity (AII), Amsterdam University Medical Centre, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Matthijs R. A. Welkers
- Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Agnes I. Veldkamp
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Marianne A. Kuijvenhoven
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - David C. de Leeuw
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Heshu Abdullah-Koolmees
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Postbus 85500, 3508 GA Utrecht, The Netherlands
- Clinical Pharmacy, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Maria T. Kuipers
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Imke H. Bartelink
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081HV Amsterdam, The Netherlands
| |
Collapse
|
17
|
Nandi R, Bhowmik D, Srivastava R, Prakash A, Kumar D. Discovering potential inhibitors against SARS-CoV-2 by targeting Nsp13 Helicase. J Biomol Struct Dyn 2022; 40:12062-12074. [PMID: 34455933 DOI: 10.1080/07391102.2021.1970024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The rise in the incidence of COVID-19 as a result of SARS-CoV-2 infection has threatened public health globally. Till now, there have been no proper prophylactics available to fight COVID-19, necessitating the advancement and evolution of effective curative against SARS-CoV-2. This study aimed at the nonstructural protein 13 (nsp13) helicase as a promising target for drug development against COVID-19. A unique collection of nucleoside analogs was screened against the SARS-CoV-2 helicase protein, for which a molecular docking experiment was executed to depict the selected ligand's binding affinity with the SARS-CoV-2 helicase proteins. Simultaneously, molecular dynamic simulations were performed to examine the protein's binding site's conformational stability, flexibility, and interaction with the ligands. Key nucleoside ligands were selected for pharmacokinetic analysis based on their docking scores. Selected ligands (cordycepin and pritelivir) showed excellent pharmacokinetics and were well stabilized at the proteins' binding site throughout the MD simulation. We have also performed binding free energy analysis or the binding characteristics of ligands with Nsp13 by using MM-PBSA and MM-GBSA. Free energy calculation by MM-PBSA and MM-GBSA analysis suggests that pritelivir may work as viable therapeutics for efficient drug advancement against SARS-CoV-2 Nsp13 helicase, potentially arresting the SARS-CoV-2 replication.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rajat Nandi
- Department of Microbiology, Assam University, Silchar, Assam, India
| | - Deep Bhowmik
- Department of Microbiology, Assam University, Silchar, Assam, India
| | - Rakesh Srivastava
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Amresh Prakash
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurgaon, Haryana, India
| | - Diwakar Kumar
- Department of Microbiology, Assam University, Silchar, Assam, India
| |
Collapse
|
18
|
Acyclovir resistance in herpes simplex viruses: Prevalence and therapeutic alternatives. Biochem Pharmacol 2022; 206:115322. [DOI: 10.1016/j.bcp.2022.115322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 11/19/2022]
|
19
|
Karpov DS, Demidova NA, Kulagin KA, Shuvalova AI, Kovalev MA, Simonov RA, Karpov VL, Snezhkina AV, Kudryavtseva AV, Klimova RR, Kushch AA. Complete and Prolonged Inhibition of Herpes Simplex Virus Type 1 Infection In Vitro by CRISPR/Cas9 and CRISPR/CasX Systems. Int J Mol Sci 2022; 23:ijms232314847. [PMID: 36499174 PMCID: PMC9738314 DOI: 10.3390/ijms232314847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Almost all people become infected with herpes viruses, including herpes simplex virus type 1 (HSV-1), during their lifetime. Typically, these viruses persist in a latent form that is resistant to all available antiviral medications. Under certain conditions, such as immunosuppression, the latent forms reactivate and cause disease. Moreover, strains of herpesviruses that are drug-resistant have rapidly emerged. Therefore, it is important to develop alternative methods capable of eradicating herpesvirus infections. One promising direction is the development of CRISPR/Cas systems for the therapy of herpesvirus infections. We aimed to design a CRISPR/Cas system for relatively effective long-term and safe control of HSV-1 infection. Here, we show that plasmids encoding the CRISPR/Cas9 system from Streptococcus pyogenes with a single sgRNA targeting the UL30 gene can completely suppress HSV-1 infection of the Vero cell line within 6 days and provide substantial protection within 9 days. For the first time, we show that CRISPR/CasX from Deltaproteobacteria with a single guide RNA against UL30 almost completely suppresses HSV-1 infection of the Vero cell line for 3 days and provides substantial protection for 6 days. We also found that the Cas9 protein without sgRNAs attenuates HSV-1 infection. Our results show that the developed CRISPR/Cas systems are promising therapeutic approaches to control HSV-1 infections.
Collapse
Affiliation(s)
- Dmitry S. Karpov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia
- Correspondence: ; Tel.: +7-(499)-135-98-01
| | - Natalia A. Demidova
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Gamaleya Str., 18, 123098 Moscow, Russia
| | - Kirill A. Kulagin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia
| | - Anastasija I. Shuvalova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia
| | - Maxim A. Kovalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia
| | - Ruslan A. Simonov
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Gamaleya Str., 18, 123098 Moscow, Russia
| | - Vadim L. Karpov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia
| | - Anastasiya V. Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia
| | - Anna V. Kudryavtseva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia
| | - Regina R. Klimova
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Gamaleya Str., 18, 123098 Moscow, Russia
| | - Alla A. Kushch
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Gamaleya Str., 18, 123098 Moscow, Russia
| |
Collapse
|
20
|
Birkmann A, Bonsmann S, Kropeit D, Pfaff T, Rangaraju M, Sumner M, Timmler B, Zimmermann H, Buschmann H, Ruebsamen-Schaeff H. Discovery, Chemistry, and Preclinical Development of Pritelivir, a Novel Treatment Option for Acyclovir-Resistant Herpes Simplex Virus Infections. J Med Chem 2022; 65:13614-13628. [PMID: 36202389 PMCID: PMC9620171 DOI: 10.1021/acs.jmedchem.2c00668] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
When the nucleoside analogue acyclovir was introduced
in the early
1980s, it presented a game-changing treatment modality for herpes
simplex virus infections. Since then, work has been ongoing to improve
the weaknesses that have now been identified: a narrow time window
for therapeutic success, resistance in immunocompromised patients,
little influence on frequency of recurrences, relatively fast elimination,
and poor bioavailability. The present Drug Annotation focuses on the
helicase–primase inhibitor pritelivir currently in development
for the treatment of acyclovir-resistant HSV infections and describes
how a change of the molecular target (from viral DNA polymerase to
the HSV helicase–primase complex) afforded improvement of the
shortcomings of nucleoside analogs. Details are presented for the
discovery process leading to the final drug candidate, the pivotal
preclinical studies on mechanism of action and efficacy, and on how
ongoing clinical research has been able to translate preclinical promises
into clinical use.
Collapse
Affiliation(s)
| | | | - Dirk Kropeit
- AiCuris Anti-Infective Cures AG, 42117 Wuppertal, Germany
| | - Tamara Pfaff
- AiCuris Anti-Infective Cures AG, 42117 Wuppertal, Germany
| | | | - Melanie Sumner
- AiCuris Anti-Infective Cures AG, 42117 Wuppertal, Germany
| | | | | | | | | |
Collapse
|
21
|
Labib BA, Chigbu DI. Clinical Management of Herpes Simplex Virus Keratitis. Diagnostics (Basel) 2022; 12:diagnostics12102368. [PMID: 36292060 PMCID: PMC9600940 DOI: 10.3390/diagnostics12102368] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/29/2022] Open
Abstract
Herpes simplex virus (HSV) keratitis is one of the leading causes of blindness worldwide. Additionally, up to 90% of the population in some countries is seropositive for HSV. HSV can cause a wide spectrum of ocular disease ranging from blepharitis to retinitis. Although the initial clinical expressions of HSV-1 and HSV-2 are similar, HSV-2 has been reported more frequently in association with recurrent HSV disease. Besides irreversible vision loss from keratitis, HSV also causes encephalitis and genital forms of the disease. Despite these statistics, there remains no vaccine against HSV. Current treatment therapies for related ocular diseases include the use of oral and topical antivirals and topical corticosteroids. While effective in many cases, they fail to address the latency and elimination of the virus, making it ineffective in addressing recurrences, a factor which increases the risk of vision loss. As such, there is a need for continued research of other potential therapeutic targets. This review utilized several published articles regarding the manifestations of HSV keratitis, antiviral immune responses to HSV infection, and clinical management of HSV keratitis. This review will summarize the current knowledge on the host–virus interaction in HSV infections, as well as highlighting the current and potential antiviral therapeutics.
Collapse
|
22
|
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
|
23
|
Wright LR, Wright DL, Weller SK. Viral Nucleases from Herpesviruses and Coronavirus in Recombination and Proofreading: Potential Targets for Antiviral Drug Discovery. Viruses 2022; 14:1557. [PMID: 35891537 PMCID: PMC9324378 DOI: 10.3390/v14071557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 02/05/2023] Open
Abstract
In this review, we explore recombination in two very different virus families that have become major threats to human health. The Herpesviridae are a large family of pathogenic double-stranded DNA viruses involved in a range of diseases affecting both people and animals. Coronaviridae are positive-strand RNA viruses (CoVs) that have also become major threats to global health and economic stability, especially in the last two decades. Despite many differences, such as the make-up of their genetic material (DNA vs. RNA) and overall mechanisms of genome replication, both human herpes viruses (HHVs) and CoVs have evolved to rely heavily on recombination for viral genome replication, adaptation to new hosts and evasion of host immune regulation. In this review, we will focus on the roles of three viral exonucleases: two HHV exonucleases (alkaline nuclease and PolExo) and one CoV exonuclease (ExoN). We will review the roles of these three nucleases in their respective life cycles and discuss the state of drug discovery efforts against these targets.
Collapse
Affiliation(s)
- Lee R. Wright
- Department of Pharmaceutical Sciences, University of Connecticut School of Pharmacy, Storrs, CT 06269, USA; (L.R.W.); (D.L.W.)
| | - Dennis L. Wright
- Department of Pharmaceutical Sciences, University of Connecticut School of Pharmacy, Storrs, CT 06269, USA; (L.R.W.); (D.L.W.)
| | - Sandra K. Weller
- Department of Molecular Biology and Biophysics, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| |
Collapse
|
24
|
Serris A, Pouvaret A, Loiseau C, Abid H, Burrel S, Fourgeaud J, Rouzaud C, Lanternier F, Boutolleau D, Frange P. Pritelivir for recurrent aciclovir-resistant herpes simplex virus 2 infections in immunocompromised patients. J Antimicrob Chemother 2022; 77:2303-2305. [PMID: 35639560 DOI: 10.1093/jac/dkac165] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Alexandra Serris
- Service de Maladies infectieuses et tropicales, Hôpital Necker - Enfants malades, Groupe hospitalo-universitaire Assistance Publique - Hôpitaux de Paris (APHP) Centre - Université Paris Cité, Paris, France
| | - Anne Pouvaret
- Service de Maladies infectieuses et tropicales, Hôpital Necker - Enfants malades, Groupe hospitalo-universitaire Assistance Publique - Hôpitaux de Paris (APHP) Centre - Université Paris Cité, Paris, France
| | - Clémence Loiseau
- Service d'hématologie, Hôpital Necker - Enfants malades, Groupe hospitalo-universitaire APHP Centre - Université Paris Cité, Paris, France
| | - Hanene Abid
- Laboratoire de microbiologie clinique, Hôpital Necker - Enfants malades, Groupe hospitalo-universitaire APHP Centre - Université Paris Cité, Paris, France
| | - Sonia Burrel
- Laboratoire de virologie, Centre National de Référence Herpèsvirus (laboratoire associé), Hôpital Pitié-Salpêtrière, Groupe hospitalo-universitaire APHP, Sorbonne Université, Paris, France.,Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP), INSERM, Sorbonne Université, Paris, France
| | - Jacques Fourgeaud
- Laboratoire de microbiologie clinique, Hôpital Necker - Enfants malades, Groupe hospitalo-universitaire APHP Centre - Université Paris Cité, Paris, France.,Centre National de Référence Herpèsvirus (laboratoire associé), Hôpital Necker - Enfants malades, Groupe hospitalo-universitaire APHP Centre - Université Paris Cité, Paris, France.,EHU 7328 PACT, Institut Imagine, Université Paris Cité, Paris, France
| | - Claire Rouzaud
- Service de Maladies infectieuses et tropicales, Hôpital Necker - Enfants malades, Groupe hospitalo-universitaire Assistance Publique - Hôpitaux de Paris (APHP) Centre - Université Paris Cité, Paris, France
| | - Fanny Lanternier
- Service de Maladies infectieuses et tropicales, Hôpital Necker - Enfants malades, Groupe hospitalo-universitaire Assistance Publique - Hôpitaux de Paris (APHP) Centre - Université Paris Cité, Paris, France
| | - David Boutolleau
- Laboratoire de virologie, Centre National de Référence Herpèsvirus (laboratoire associé), Hôpital Pitié-Salpêtrière, Groupe hospitalo-universitaire APHP, Sorbonne Université, Paris, France.,Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP), INSERM, Sorbonne Université, Paris, France
| | - Pierre Frange
- Laboratoire de microbiologie clinique, Hôpital Necker - Enfants malades, Groupe hospitalo-universitaire APHP Centre - Université Paris Cité, Paris, France.,EHU 7328 PACT, Institut Imagine, Université Paris Cité, Paris, France
| |
Collapse
|
25
|
Raubenolt BA, Islam NN, Summa CM, Rick SW. Molecular dynamics simulations of the flexibility and inhibition of SARS-CoV-2 NSP 13 helicase. J Mol Graph Model 2022; 112:108122. [PMID: 35021142 PMCID: PMC8730789 DOI: 10.1016/j.jmgm.2022.108122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 11/25/2022]
Abstract
The helicase protein of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is both a good potential drug target and very flexible. The flexibility, and therefore its function, could be reduced through knowledge of these motions and identification of allosteric pockets. Using molecular dynamics simulations with enhanced sampling, we determined key modes of motion and sites on the protein that are at the interface between flexible domains of the proteins. We developed an approach to map the principal components of motion onto the surface of a potential binding pocket to help in the identification of allosteric sites.
Collapse
Affiliation(s)
- Bryan A Raubenolt
- Department of Chemistry, University of New Orleans, New Orleans, LA, 70148, USA
| | - Naeyma N Islam
- Department of Chemistry, University of New Orleans, New Orleans, LA, 70148, USA
| | - Christoper M Summa
- Department of Computer Science, University of New Orleans, New Orleans, LA, 70148, USA
| | - Steven W Rick
- Department of Chemistry, University of New Orleans, New Orleans, LA, 70148, USA.
| |
Collapse
|
26
|
Spahn S, Riessen R, Berg CP, Malek NP, Emrich MH, Lohrengel K, Ganzenmueller T, Iftner T, Kleymann G, Hamprecht K. Comprehensive clinical and virological characterization of three cases of fulminant liver failure owing to HSV1 primary infection. Liver Int 2022; 42:1005-1011. [PMID: 35230726 DOI: 10.1111/liv.15215] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/23/2021] [Accepted: 01/16/2022] [Indexed: 12/13/2022]
Abstract
Herpes simplex virus 1 (HSV-1) is a frequently unrecognized, yet deadly cause of acute liver failure (ALF). We, therefore, analysed three cases of fatal HSV-1-induced ALF. All patients shared clinical (extremely elevated transaminases, LDH and AST/LDH ratio < 1) and virological characteristics (ratio of viral load in plasma versus throat swabs: 60-700-fold, lack of anti-HSV-1-IgG antibodies or low IgG-avidity during primary infection), which may help to identify patients at risk. Additionally, in vitro chemosusceptibility assays revealed high efficacy of the helicase-primase inhibitors (HPI), pritelivir and drug-candidate IM-250 compared to acyclovir (ACV) using HSV-1-isolates from two patients; hence, ACV/HPI-combinations might offer new therapeutic options for HSV-induced ALF.
Collapse
Affiliation(s)
- Stephan Spahn
- Department Internal Medicine I, University Hospital of Tuebingen, Tuebingen, Germany
| | - Reimer Riessen
- Department of Internal Medicine, Medical Intensive Care Unit, University Hospital of Tuebingen, Tübingen, Germany
| | - Christoph P Berg
- Department Internal Medicine I, University Hospital of Tuebingen, Tuebingen, Germany
| | - Nisar P Malek
- Department Internal Medicine I, University Hospital of Tuebingen, Tuebingen, Germany
| | - Marzena H Emrich
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital of Tuebingen, Tübingen, Germany
| | - Kristina Lohrengel
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital of Tuebingen, Tübingen, Germany
| | - Tina Ganzenmueller
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital of Tuebingen, Tübingen, Germany
| | - Thomas Iftner
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital of Tuebingen, Tübingen, Germany
| | - Gerald Kleymann
- University of Tübingen IFIB, Interfaculty Institute for Biochemistry, Tübingen, Germany.,Innovative Molecules GmbH, Bad-Salzuflen, Germany
| | - Klaus Hamprecht
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital of Tuebingen, Tübingen, Germany
| |
Collapse
|
27
|
Johnston C. Diagnosis and Management of Genital Herpes: Key Questions and Review of the Evidence for the 2021 Centers for Disease Control and Prevention Sexually Transmitted Infections Treatment Guidelines. Clin Infect Dis 2022; 74:S134-S143. [PMID: 35416970 DOI: 10.1093/cid/ciab1056] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Genital herpes, caused by herpes simplex virus (HSV) type 1 or type 2, is a prevalent sexually transmitted infection (STI). Given that HSV is an incurable infection, there are important concerns about appropriate use of diagnostic tools, management of infection, prevention of transmission to sexual partners, and appropriate counseling. In preparation for updating the Centers for Disease Control and Prevention (CDC) STI treatment guidelines, key questions for management of genital herpes infection were developed with a panel of experts. To answer these questions, a systematic literature review was performed, with tables of evidence including articles that would change guidance assembled. These data were used to inform recommendations in the 2021 CDC STI treatment guidelines.
Collapse
Affiliation(s)
- Christine Johnston
- Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.,University of Washington, Seattle, Washington, USA.,Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
28
|
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
|
29
|
Cheng H, Yang T, Edwards M, Tang S, Xu S, Yan X. Picomole-Scale Transition Metal Electrocatalysis Screening Platform for Discovery of Mild C-C Coupling and C-H Arylation through in Situ Anodically Generated Cationic Pd. J Am Chem Soc 2022; 144:1306-1312. [PMID: 35015550 DOI: 10.1021/jacs.1c11179] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Development of new transition-metal-catalyzed electrochemistry promises to improve overall synthetic efficiency. Here, we describe the first integrated platform for online screening of electrochemical transition-metal catalysis. It utilizes the intrinsic electrochemical capabilities of nanoelectrospray ionization mass spectrometry (nano-ESI-MS) and picomole-scale anodic corrosion of a Pd electrode to generate and evaluate highly efficient cationic catalysts for mild electrocatalysis. We demonstrate the power of the novel electrocatalysis platform by (1) identifying electrolytic Pd-catalyzed Suzuki coupling at room temperature, (2) discovering Pd-catalyzed electrochemical C-H arylation in the absence of external oxidant or additive, (3) developing electrolyzed Suzuki coupling/C-H arylation cascades, and (4) achieving late-stage functionalization of two drug molecules by the newly developed mild electrocatalytic C-H arylation. More importantly, the scale-up reactions confirm that new electrochemical pathways discovered by nano-ESI can be implemented under the conventional electrolytic reaction conditions. This approach enables in situ mechanistic studies by capturing various intermediates including transient transition metal species by MS, and thus uncovering the critical role of anodically generated cationic Pd catalyst in promoting otherwise sluggish transmetalation in C-H arylation. The anodically generated cationic Pd with superior catalytic efficiency and novel online electrochemical screening platform hold great potential for discovering mild transition-metal-catalyzed reactions.
Collapse
Affiliation(s)
- Heyong Cheng
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States.,College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China
| | - Tingyuan Yang
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Madison Edwards
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Shuli Tang
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Shiqing Xu
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Xin Yan
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
30
|
|
31
|
Romero N, Favoreel HW. Pseudorabies Virus Infection Triggers NF-κB Activation via the DNA Damage Response but Actively Inhibits NF-κB-Dependent Gene Expression. J Virol 2021; 95:e0166621. [PMID: 34613805 PMCID: PMC8610585 DOI: 10.1128/jvi.01666-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/01/2021] [Indexed: 12/22/2022] Open
Abstract
The nuclear factor kappa B (NF-κB) pathway is known to integrate signaling associated with very diverse intra- and extracellular stressors, including virus infections, and triggers a powerful (proinflammatory) response through the expression of NF-κB-regulated genes. Typically, the NF-κB pathway collects and transduces threatening signals at the cell surface or in the cytoplasm leading to nuclear import of activated NF-κB transcription factors. In the current work, we demonstrate that the swine alphaherpesvirus pseudorabies virus (PRV) induces a peculiar mode of NF-κB activation known as "inside-out" NF-κB activation. We show that PRV triggers the DNA damage response (DDR) and that this DDR response drives NF-κB activation since inhibition of the nuclear ataxia telangiectasia-mutated (ATM) kinase, a chief controller of DDR, abolished PRV-induced NF-κB activation. Initiation of the DDR-NF-κB signaling axis requires viral protein synthesis but occurs before active viral genome replication. In addition, the initiation of the DDR-NF-κB signaling axis is followed by a virus-induced complete shutoff of NF-κB-dependent gene expression that depends on viral DNA replication. In summary, the results presented in this study reveal that PRV infection triggers a noncanonical DDR-NF-κB activation signaling axis and that the virus actively inhibits the (potentially antiviral) consequences of this pathway, by inhibiting NF-κB-dependent gene expression. IMPORTANCE The NF-κB signaling pathway plays a critical role in coordination of innate immune responses that are of vital importance in the control of infections. The current report generates new insights into the interaction of the alphaherpesvirus pseudorabies virus (PRV) with the NF-κB pathway, as they reveal that (i) PRV infection leads to NF-κB activation via a peculiar "inside-out" nucleus-to-cytoplasm signal that is triggered via the DNA damage response (DDR), (ii) the DDR-NF-κB signaling axis requires expression of viral proteins but is initiated before active PRV replication, and (iii) late viral factor(s) allow PRV to actively and efficiently inhibit NF-κB-dependent (proinflammatory) gene expression. These data suggest that activation of the DDR-NF-κB during PRV infection is host driven and that its potential antiviral consequences are actively inhibited by the virus.
Collapse
Affiliation(s)
- Nicolás Romero
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Herman W. Favoreel
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
32
|
Amenamevir, a Helicase-Primase Inhibitor, for the Optimal Treatment of Herpes Zoster. Viruses 2021; 13:v13081547. [PMID: 34452412 PMCID: PMC8402822 DOI: 10.3390/v13081547] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 12/02/2022] Open
Abstract
Acyclovir, valacyclovir, and famciclovir are used for the treatment of herpes simplex virus (HSV) and varicella-zoster virus (VZV) infections. Helicase-primase inhibitors (HPIs) inhibit replication fork progression that separates double DNA strands into two single strands during DNA synthesis. The HPIs amenamevir and pritelivir have novel mechanisms of anti-herpetic action, and their once-daily administration has clinical efficacy for genital herpes. Among HPIs, amenamevir has anti-VZV activity. The concentrations of HSV-1 and VZV required for the 50% plaque reduction of amenamevir were 0.036 and 0.047 μM, respectively. We characterized the features of amenamevir regarding its mechanism, resistance, and synergism with acyclovir. Its antiviral activity was not influenced by the viral replication cycle, in contrast to acyclovir. A clinical trial of amenamevir for herpes zoster demonstrated its non-inferiority to valacyclovir. To date, amenamevir has been successfully used in over 1,240,000 patients with herpes zoster in Japan. Post-marketing surveillance of amenamevir in Japan reported side effects with significant potential risk identified by the Japanese Risk Management Plan, including thrombocytopenia, gingival bleeding, and palpitations, although none of these were serious. The clinical efficacy and safety profiles of amenamevir were established in patients with herpes zoster. Therefore, amenamevir as an HPI opens a new era of anti-herpes therapy.
Collapse
|
33
|
Gege C, Bravo FJ, Uhlig N, Hagmaier T, Schmachtenberg R, Elis J, Burger-Kentischer A, Finkelmeier D, Hamprecht K, Grunwald T, Bernstein DI, Kleymann G. A helicase-primase drug candidate with sufficient target tissue exposure affects latent neural herpes simplex virus infections. Sci Transl Med 2021; 13:13/598/eabf8668. [PMID: 34135112 DOI: 10.1126/scitranslmed.abf8668] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/26/2021] [Indexed: 12/17/2022]
Abstract
More than 50% of the world population is chronically infected with herpesviruses. Herpes simplex virus (HSV) infections are the cause of herpes labialis (cold sores), genital herpes, and sight-impairing keratitis. Less frequently, life-threatening disseminated disease (encephalitis and generalized viremia) can also occur, mainly in immunocompromised patients and newborns. After primary infection, HSV persists for life in a latent state in trigeminal or sacral ganglia and, triggered by diverse stimuli, disease recurs in more than 30% of patients up to several times a year. Current therapy with nucleoside analogs targeting the viral polymerase is somewhat effective but limited by poor exposure in the nervous system, and latent infections are not affected by therapy. Here, we report on an inhibitor of HSV helicase-primase with potent in vitro anti-herpes activity, a different mechanism of action, a low frequency of HSV resistance, and a favorable pharmacokinetic and safety profile. Improved target tissue exposure results in superior efficacy in preventing and treating HSV infection and disease in animal models as compared to standard of care. Therapy of primary HSV infections with drug candidate IM-250 {(S)-2-(2',5'-difluoro-[1,1'-biphenyl]-4-yl)-N-methyl-N-(4-methyl-5-(S-methylsulfon-imidoyl)thiazol-2-yl)acetamide} not only reduces the duration of disease symptoms or time to healing but also prevents recurrent disease in guinea pigs. Treatment of recurrent infections reduces the frequency of recurrences and viral shedding, and, unlike nucleosidic drugs, IM-250 remains effective for a time after cessation of treatment. Hence, IM-250 has advantages over standard-of-care therapies and represents a promising therapeutic for chronic HSV infection, including nucleoside-resistant HSV.
Collapse
Affiliation(s)
- Christian Gege
- Innovative Molecules GmbH, Leopoldshöher Str. 7, 32107 Bad-Salzuflen, Germany
| | - Fernando J Bravo
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center CCHMC, 3333 Burnet Ave., Cincinnati, OH 45229, USA
| | - Nadja Uhlig
- Fraunhofer IZI, Institute for Cell Therapy and Immunology, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Timo Hagmaier
- University of Tübingen IFIB, Interfaculty Institute for Biochemistry, Auf der Morgenstelle 34, 72076 Tübingen, Germany
| | | | - Julia Elis
- University of Tübingen IFIB, Interfaculty Institute for Biochemistry, Auf der Morgenstelle 34, 72076 Tübingen, Germany
| | - Anke Burger-Kentischer
- Fraunhofer IGB, Institute for Interfacial Engineering and Biotechnology, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Doris Finkelmeier
- Fraunhofer IGB, Institute for Interfacial Engineering and Biotechnology, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Klaus Hamprecht
- University Hospital of Tübingen UKT, Institute for Medical Virology and Epidemiology of Viral Diseases, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| | - Thomas Grunwald
- Fraunhofer IZI, Institute for Cell Therapy and Immunology, Perlickstrasse 1, 04103 Leipzig, Germany
| | - David I Bernstein
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center CCHMC, 3333 Burnet Ave., Cincinnati, OH 45229, USA
| | - Gerald Kleymann
- Innovative Molecules GmbH, Leopoldshöher Str. 7, 32107 Bad-Salzuflen, Germany. .,University of Tübingen IFIB, Interfaculty Institute for Biochemistry, Auf der Morgenstelle 34, 72076 Tübingen, Germany
| |
Collapse
|
34
|
Kim J, Park SJ, Park J, Shin H, Jang YS, Woo JS, Min DH. Identification of a Direct-Acting Antiviral Agent Targeting RNA Helicase via a Graphene Oxide Nanobiosensor. ACS APPLIED MATERIALS & INTERFACES 2021; 13:25715-25726. [PMID: 34036784 DOI: 10.1021/acsami.1c04641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Dengue virus (DENV), an arbovirus transmitted by mosquitoes, causes infectious diseases such as dengue fever, dengue hemorrhagic fever, and dengue shock syndrome. Despite the dangers posed by DENV, there are no approved antiviral drugs for treatment of DENV infection. Considering the potential for a global dengue outbreak, rapid development of antiviral agents against DENV infections is crucial as a preemptive measure; thus, the selection of apparent drug targets, such as the viral enzymes involved in the viral life cycle, is recommended. Helicase, a potential drug target in DENV, is a crucial viral enzyme that unwinds double-stranded viral RNA, releasing single-stranded RNA genomes during viral replication. Therefore, an inhibitor of helicase activity could serve as a direct-acting antiviral agent. Here, we introduce an RNA helicase assay based on graphene oxide, which enables fluorescence-based analysis of RNA substrate-specific helicase enzyme activity. This assay demonstrated high reliability and ability for high-throughput screening, identifying a new helicase inhibitor candidate, micafungin (MCFG), from an FDA-approved drug library. As a direct-acting antiviral agent targeting RNA helicase, MCFG inhibits DENV proliferation in cells and an animal model. Notably, in vivo, MCFG treatment reduced viremia, inflammatory cytokine levels, and viral loads in several tissues and improved survival rates by up to 40% in a lethal mouse model. Therefore, we suggest MCFG as a potential direct-acting antiviral drug candidate.
Collapse
Affiliation(s)
- Jungho Kim
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Se-Jin Park
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Jisang Park
- Department of Bioactive Material Sciences and Institute of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Hojeong Shin
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Yong-Suk Jang
- Department of Bioactive Material Sciences and Institute of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Jae-Sung Woo
- Center for RNA Research, Institute for Basic Science (IBS), Seoul National University, Seoul 08826, Republic of Korea
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Biotherapeutics Convergence Technology, Lemonex Inc., Seoul 06683, Republic of Korea
| |
Collapse
|
35
|
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
|
36
|
Samies NL, James SH, Kimberlin DW. Neonatal Herpes Simplex Virus Disease: Updates and Continued Challenges. Clin Perinatol 2021; 48:263-274. [PMID: 34030813 DOI: 10.1016/j.clp.2021.03.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
This article defines neonatal herpes simplex virus (HSV) disease and describes the progress over the past 40 years that has revolutionized the management of HSV disease in neonates to improve their outcomes. These advancements include the introduction of acyclovir in the 1980s, polymerase chain reaction (PCR) for the detection of HSV DNA in the 1990s, and recommendations on managing infants born to mothers with active genital lesions. Despite these advancements, however, there remain high morbidity and mortality in affected neonates, with need for continued improvement. Areas of high interest include vaccine development and rapid PCR detection at time of delivery.
Collapse
Affiliation(s)
- Nicole L Samies
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Alabama at Birmingham, Children's Harbor Building 308, 1600 7th Avenue South, Birmingham, AL 35233-1711, USA.
| | - Scott H James
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Alabama at Birmingham, Children's Harbor Building 308, 1600 7th Avenue South, Birmingham, AL 35233-1711, USA
| | - David W Kimberlin
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Alabama at Birmingham, Children's Harbor Building 308, 1600 7th Avenue South, Birmingham, AL 35233-1711, USA
| |
Collapse
|
37
|
Cannon L, Tholouli E, Ward C, Farooq H, Kingston M. Use of pritelivir in refractory aciclovir-resistant herpes simplex virus type 2. Int J STD AIDS 2021; 32:978-980. [PMID: 33947276 DOI: 10.1177/09564624211006568] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recurrence of mucocutaneous herpes simplex virus (HSV) infections is common in immunosuppressed patients. Thymidine kinase mutations conferring resistance to the antiviral agent aciclovir have been observed in such patients. Recommended second-line therapeutic agents against HSV are associated with significant side effects contributing to disease burden. We present a case of aciclovir-resistant herpes simplex virus 2 (HSV-2) in an immunosuppressed (HIV negative) allogenic peripheral blood stem cell transplant (SCT) recipient which was refractory to second-line therapy. Compassionate acquisition of the novel oral helicase-primase inhibitor pritelivir provided both symptomatic and virological control for the duration of its use. We believe this to be the first clinical use of this therapeutic agent in the United Kingdom.
Collapse
Affiliation(s)
- Luke Cannon
- Northern Sexual Health, 8943Contraception and HIV Service, Manchester University NHS Foundation Trust, Manchester, UK
| | - Eleni Tholouli
- Department of Haematology, 5293Manchester University NHS Foundation Trust, Manchester, UK
| | - Chris Ward
- Northern Sexual Health, 8943Contraception and HIV Service, Manchester University NHS Foundation Trust, Manchester, UK
| | | | - Margaret Kingston
- Northern Sexual Health, 8943Contraception and HIV Service, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
38
|
Shiraki K, Takemoto M, Daikoku T. Emergence of varicella-zoster virus resistance to acyclovir: epidemiology, prevention, and treatment. Expert Rev Anti Infect Ther 2021; 19:1415-1425. [PMID: 33853490 DOI: 10.1080/14787210.2021.1917992] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Acyclovir has led to the development of successful systemic therapy for herpes simplex virus and varicella-zoster virus (VZV) infection, and the use of valacyclovir and famciclovir has improved treatment. Additionally, the use of a helicase-primase (HP) inhibitor (HPI), amenamevir, is changing the treatment of herpes zoster (HZ).Area covered: VZV infection is prevented by vaccines and is treated with antiviral agents. Acyclovir and penciclovir are phosphorylated by viral thymidine kinase and work as chain terminators. Improvements in the management of immunocompromised patients have reduced severe and prolonged immunosuppression and chronic VZV infection with acyclovir-resistant mutants has become rarer. The HP is involved in the initial step of DNA synthesis and amenamevir has novel mechanisms of action, efficacy to acyclovir-resistant mutants, and pharmacokinetic characteristics. The literature search for PUBMED was conducted on 10 April 2020 and updated on 4 November 2020.Expert opinion: Amenamevir has been used to treat HZ in Japan. Although the number of patients with VZV infection will decrease owing to the use of vaccines, the addition of HPI will improve treatment and treatment options for resistant viruses. The clinical use of HPIs in addition to current nucleoside analogs opens a new era of antiherpes therapy.
Collapse
Affiliation(s)
- Kimiyasu Shiraki
- Senri Kinran University & Department of Virology, University of Toyama, Toyama, Japan
| | - Masaya Takemoto
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, Japan
| | - Tohru Daikoku
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, Japan
| |
Collapse
|
39
|
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
|
40
|
Abstract
Herpesviruses such as herpes simplex virus (HSV) type 1 and 2, varicella-zoster virus (VZV), and cytomegalovirus (CMV) maintain lifelong latency in the host after primary infection and can reactivate periodically either as asymptomatic viral shedding or as clinical disease. Immunosuppression, including biologic therapy, may increase frequency and severity of herpesvirus reactivation and infection. Licensed biologics are reviewed regarding their risks of potentiating HSV, VZV, and CMV reactivation and infection. Approaches to prophylaxis against HSV, VZV, and CMV infection or reactivation are discussed.
Collapse
Affiliation(s)
- Dora Y Ho
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Lane Building L-135, Stanford, CA 94305-5107, USA.
| | - Kyle Enriquez
- Stanford University, 450 Serra Mall, Stanford, CA 94305, USA
| | - Ashrit Multani
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue CHS 37-121, Los Angeles, CA 90095-1688, USA
| |
Collapse
|
41
|
Piret J, Boivin G. Antiviral Drugs Against Herpesviruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1322:1-30. [PMID: 34258735 DOI: 10.1007/978-981-16-0267-2_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The discovery of the nucleoside analogue, acyclovir, represented a milestone in the management of infections caused by herpes simplex virus and varicella-zoster virus. Ganciclovir, another nucleoside analogue, was then used for the management of systemic and organ-specific human cytomegalovirus diseases. The pyrophosphate analogue, foscarnet, and the nucleotide analogue, cidofovir, have been approved subsequently and constitute the second-line antiviral drugs. However, the viral DNA polymerase is the ultimate target of all these antiviral agents with a possible emergence of cross-resistance between these drugs. Recently, letermovir that targets the viral terminase complex was approved for the prophylaxis of human cytomegalovirus infections in hematopoietic stem cell transplant recipients. Other viral targets such as the protein kinase and the helicase-primase complex are also evaluated for the development of novel potent inhibitors against herpesviruses.
Collapse
Affiliation(s)
| | - Guy Boivin
- CHU de Québec-Laval University, Quebec City, QC, Canada.
| |
Collapse
|
42
|
Castillo E, Duarte LF, Corrales N, Álvarez DM, Farías MA, Henríquez A, Smith PC, Agurto-Muñoz C, González PA. Anti-herpetic Activity of Macrocystis pyrifera and Durvillaea antarctica Algae Extracts Against HSV-1 and HSV-2. Front Microbiol 2020; 11:2006. [PMID: 33013743 PMCID: PMC7516053 DOI: 10.3389/fmicb.2020.02006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/29/2020] [Indexed: 12/21/2022] Open
Abstract
Herpes simplex viruses (HSVs) type 1 (HSV-1) and type 2 (HSV-2) are highly prevalent in the human population, and the infections they produce are lifelong with frequent reactivations throughout life. Both viruses produce uncomfortable and sometimes painful lesions in the orofacial and genital areas, as well as herpetic gingivostomatitis, among other clinical manifestations. At present, the most common treatments against HSVs consist of nucleoside analogs that target the viral polymerases. However, such drugs are poorly effective for treating skin lesions, as they only reduce in 1-2 days the duration of the herpetic lesions. Additionally, viral isolates resistant to these drugs can emerge in immunosuppressed individuals, and second-line drugs for such variants are frequently accompanied by adverse effects requiring medical supervision. Thus, novel or improved therapeutic drugs for treating HSV lesions are needed. Here, we assessed the potential antiviral activity of aqueous extracts obtained from two brown macroalgae, namely Macrocystis pyrifera and Durvillaea antarctica against HSVs. Both extracts showed antiviral activity against acyclovir-sensitive and acyclovir-resistant HSV-1 and HSV-2. Our analyses show that there is a significant antiviral activity associated with proteins in the extract, although other compounds also seem to contribute to inhibiting the replication cycle of these viruses. Evaluation of the algae extracts as topical formulations in an animal model of HSV-1 skin infection significantly reduced the severity of the disease more than acyclovir, as well as the duration of the herpetic lesions, when compared to mock-treated animals, with the D. antarctica extract performing best. Taken together, these findings suggest that these algae extracts may be potential phytotherapeutics against HSVs and may be useful for the treatment and reduction of common herpetic manifestations in humans.
Collapse
Affiliation(s)
- Estefanía Castillo
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luisa F. Duarte
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolas Corrales
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Diana M. Álvarez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A. Farías
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Adolfo Henríquez
- GIBMAR, Grupo Interdisciplinario de Biotecnología Marina, Centro de Biotecnología, Universidad de Concepción, Concepción, Chile
| | - Patricio C. Smith
- Escuela de Odontología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian Agurto-Muñoz
- GIBMAR, Grupo Interdisciplinario de Biotecnología Marina, Centro de Biotecnología, Universidad de Concepción, Concepción, Chile
- Departamento de Ciencia y Tecnología de los Alimentos, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
43
|
Sibley D, Larkin DFP. Update on Herpes simplex keratitis management. Eye (Lond) 2020; 34:2219-2226. [PMID: 32843744 DOI: 10.1038/s41433-020-01153-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/14/2020] [Accepted: 08/14/2020] [Indexed: 12/21/2022] Open
Abstract
Herpes simplex keratitis (HSK) is a common, potentially blinding condition characterised by recurrent infections of the cornea, seen by both general ophthalmologists and corneal specialists. Successful treatment of recurrences reduces disease duration, prevents progressive corneal scaring leading to vision loss and reduces risk of further recurrences. In this review we discuss the relative advantages of the diagnostic laboratory investigations including polymerase chain reaction, viral culture and fluorescence-based immunohistochemistry. We review treatment strategies in selected aspects of HSK and discuss the management options in cases not responding to treatment.
Collapse
Affiliation(s)
- Daniel Sibley
- Cornea & External Diseases Service, Moorfields Eye Hospital, 162 City Road, London, EC1V 2PD, UK
| | - Daniel F P Larkin
- Cornea & External Diseases Service, Moorfields Eye Hospital, 162 City Road, London, EC1V 2PD, UK. .,NIHR Moorfields Clinical Research Facility, Moorfields Eye Hospital, London, UK. .,UCL Institute of Ophthalmology, London, UK.
| |
Collapse
|
44
|
Anton-Vazquez V, Mehra V, Mbisa JL, Bradshaw D, Basu TN, Daly ML, Mufti GJ, Pagliuca A, Potter V, Zuckerman M. Challenges of aciclovir-resistant HSV infection in allogeneic bone marrow transplant recipients. J Clin Virol 2020; 128:104421. [DOI: 10.1016/j.jcv.2020.104421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/22/2020] [Accepted: 05/03/2020] [Indexed: 11/27/2022]
|
45
|
Looker KJ, Johnston C, Welton NJ, James C, Vickerman P, Turner KME, Boily MC, Gottlieb SL. The global and regional burden of genital ulcer disease due to herpes simplex virus: a natural history modelling study. BMJ Glob Health 2020; 5:e001875. [PMID: 32201620 PMCID: PMC7061890 DOI: 10.1136/bmjgh-2019-001875] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/24/2019] [Accepted: 11/10/2019] [Indexed: 11/03/2022] Open
Abstract
Introduction Herpes simplex virus (HSV) infection can cause painful, recurrent genital ulcer disease (GUD), which can have a substantial impact on sexual and reproductive health. HSV-related GUD is most often due to HSV type 2 (HSV-2), but may also be due to genital HSV type 1 (HSV-1), which has less frequent recurrent episodes than HSV-2. The global burden of GUD has never been quantified. Here we present the first global and regional estimates of GUD due to HSV-1 and HSV-2 among women and men aged 15-49 years old. Methods We developed a natural history model reflecting the clinical course of GUD following HSV-2 and genital HSV-1 infection, informed by a literature search for data on model parameters. We considered both diagnosed and undiagnosed symptomatic infection. This model was then applied to existing infection estimates and population sizes for 2016. A sensitivity analysis was carried out varying the assumptions made. Results We estimated that 187 million people aged 15-49 years had at least one episode of HSV-related GUD globally in 2016: 5.0% of the world's population. Of these, 178 million (95% of those with HSV-related GUD) had HSV-2 compared with 9 million (5%) with HSV-1. GUD burden was highest in Africa, and approximately double in women compared with men. Altogether there were an estimated 8 billion person-days spent with HSV-related GUD globally in 2016, with 99% of days due to HSV-2. Taking into account parameter uncertainty, the percentage with at least one episode of HSV-related GUD ranged from 3.2% to 7.9% (120-296 million). However, the estimates were sensitive to the model assumptions. Conclusion Our study represents a first attempt to quantify the global burden of HSV-related GUD, which is large. New interventions such as HSV vaccines, antivirals or microbicides have the potential to improve the quality of life of millions of people worldwide.
Collapse
Affiliation(s)
- Katharine Jane Looker
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Christine Johnston
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Virology Research Clinic, Seattle, Washington, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Nicky J Welton
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Charlotte James
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Peter Vickerman
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | | | - Marie-Claude Boily
- Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Sami L Gottlieb
- Department of Reproductive Health and Research, World Health Organization, Geneve, Switzerland
| |
Collapse
|
46
|
Álvarez DM, Castillo E, Duarte LF, Arriagada J, Corrales N, Farías MA, Henríquez A, Agurto-Muñoz C, González PA. Current Antivirals and Novel Botanical Molecules Interfering With Herpes Simplex Virus Infection. Front Microbiol 2020; 11:139. [PMID: 32117158 PMCID: PMC7026011 DOI: 10.3389/fmicb.2020.00139] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/21/2020] [Indexed: 12/31/2022] Open
Abstract
Herpes simplex viruses type 1 (HSV-1) and type 2 (HSV-2) are highly prevalent within the human population and are characterized by lifelong infections and sporadic recurrences due to latent neuron infection. Upon reactivations, HSVs may manifest either, symptomatically or asymptomatically and be shed onto others through mucosae body fluids. Although, HSVs can produce severe disease in humans, such as life-threatening encephalitis and blindness, the most common symptoms are skin and mucosal lesions in the oro-facial and the genital areas. Nucleoside analogs with antiviral activity can prevent severe HSV infection, yet they are not very effective for treating skin manifestations produced by these viruses, as they only reduce in a few days at most the duration of lesions. Additionally, HSV variants that are resistant to these antivirals may arise, especially in immunosuppressed individuals. Thus, new antivirals that can reduce the severity and duration of these cutaneous manifestations would certainly be welcome. Here, we review currently available anti-herpetic therapies, novel molecules being assessed in clinical trials and new botanical compounds reported in the last 20 years with antiviral activities against HSVs that might represent future treatments against these viruses.
Collapse
Affiliation(s)
- Diana M. Álvarez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Estefanía Castillo
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luisa F. Duarte
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José Arriagada
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás Corrales
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A. Farías
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Adolfo Henríquez
- Centro de Biotecnología, Universidad de Concepción, Concepción, Chile
| | - Cristian Agurto-Muñoz
- Centro de Biotecnología, Universidad de Concepción, Concepción, Chile
- Departamento de Ciencia y Tecnología de Alimentos, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
47
|
Mei C, Yang W, Wei X, Wu K, Huang D. The Unique Microbiome and Innate Immunity During Pregnancy. Front Immunol 2019; 10:2886. [PMID: 31921149 PMCID: PMC6929482 DOI: 10.3389/fimmu.2019.02886] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/25/2019] [Indexed: 12/26/2022] Open
Abstract
A successful pregnancy depends on not only the tolerance of the fetal immune system by the mother but also resistance against the threat of hazardous microorganisms. Infection with pathogenic microorganisms during pregnancy may lead to premature delivery, miscarriage, growth restriction, neonatal morbidity, and other adverse outcomes. Moreover, the host also has an intact immune system to avoid these adverse outcomes. It is important to note the presence of normal bacteria in the maternal reproductive tract and the principal role of the maternal-placental-fetal interaction in antimicrobial immunity. Previous studies mainly focused on maternal infection during pregnancy. However, this review summarizes the new views on the study of the maternal microbiome and expounds the innate immune defense mechanism of the maternal vagina and decidua as well as how cytotrophoblasts and syncytiotrophoblasts recognize and kill bacteria in the placenta. Fetal immune systems, thought to be weak, also exhibit an immune defense function that is indispensable for maintaining the safety of the fetus. The skin, lungs, and intestines of the fetus during pregnancy constitute the main immune barriers. These findings will provide a new understanding of the effects of normal microbial flora and how the host resists harmful microbes during pregnancy. We believe that it may also contribute to the reference on the clinical prevention and treatment of gestational infection to avoid adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Chunlei Mei
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weina Yang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Wei
- Second Affiliated Hospital of Jinlin University, Changchun, China
| | - Kejia Wu
- Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Donghui Huang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
48
|
Yuan WY, Chen X, Liu NN, Wen YN, Yang B, Andrei G, Snoeck R, Xiang YH, Wu YW, Jiang Z, Schols D, Zhang ZY, Wu QP. Synthesis, Anti-Varicella-Zoster Virus and Anti-Cytomegalovirus Activity of 4,5-Disubstituted 1,2,3-(1H)-Triazoles. Med Chem 2019; 15:801-812. [PMID: 30411688 DOI: 10.2174/1573406414666181109095239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 10/05/2018] [Accepted: 10/30/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Clinical drugs for herpesvirus exhibit high toxicity and suffer from significant drug resistance. The development of new, effective, and safe anti-herpesvirus agents with different mechanisms of action is greatly required. OBJECTIVE Novel inhibitors against herpesvirus with different mechanisms of action from that of clinical drugs. METHODS A series of novel 5-(benzylamino)-1H-1,2,3-triazole-4-carboxamides were efficiently synthesized and EC50 values against Human Cytomegalovirus (HCMV), Varicella-Zoster Virus (VZV) and Herpes Simplex Virus (HSV) were evaluated in vitro. RESULTS Some compounds present antiviral activity. Compounds 5s and 5t are potent against both HCMV and VZV. Compounds 5m, 5n, 5s, and 5t show similar EC50 values against both TK+ and TK- VZV strains. CONCLUSION 5-(Benzylamino)-1H-1, 2,3-triazole-4-carboxamides are active against herpesviruses and their activity is remarkably affected by the nature and the position of substituents in the benzene ring. The results indicate that these derivatives are independent of the viral thymidine kinase (TK) for activation, which is indispensable for current drugs. Their mechanisms of action may differ from those of the clinic anti-herpesvirus drugs.
Collapse
Affiliation(s)
- Wei-Yuan Yuan
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xue Chen
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Ning-Ning Liu
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Yi-Ning Wen
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Bei Yang
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Graciela Andrei
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, Leuven B-3000, Belgium
| | - Robert Snoeck
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, Leuven B-3000, Belgium
| | - Yu-Hong Xiang
- School of Chemistry, Capital Normal University, Beijing 100048, China
| | - Yong-Wei Wu
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Zhen Jiang
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Dominique Schols
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, Leuven B-3000, Belgium
| | - Zhuo-Yong Zhang
- School of Chemistry, Capital Normal University, Beijing 100048, China
| | - Qin-Pei Wu
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
49
|
Schiffer JT, Gottlieb SL. Biologic interactions between HSV-2 and HIV-1 and possible implications for HSV vaccine development. Vaccine 2019; 37:7363-7371. [PMID: 28958807 PMCID: PMC5867191 DOI: 10.1016/j.vaccine.2017.09.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 09/08/2017] [Indexed: 12/14/2022]
Abstract
Development of a safe and effective vaccine against herpes simplex virus type 2 (HSV-2) has the potential to limit the global burden of HSV-2 infection and disease, including genital ulcer disease and neonatal herpes, and is a global sexual and reproductive health priority. Another important potential benefit of an HSV-2 vaccine would be to decrease HIV infections, as HSV-2 increases the risk of HIV-1 acquisition several-fold. Acute and chronic HSV-2 infection creates ulcerations and draws dendritic cells and activated CD4+ T cells into genital mucosa. These cells are targets for HIV entry and replication. Prophylactic HSV-2 vaccines (to prevent infection) and therapeutic vaccines (to modify or treat existing infections) are currently under development. By preventing or modifying infection, an effective HSV-2 vaccine could limit HSV-associated genital mucosal inflammation and thus HIV risk. However, a vaccine might have competing effects on HIV risk depending on its mechanism of action and cell populations generated in the genital mucosa. In this article, we review biologic interactions between HSV-2 and HIV-1, consider HSV-2 vaccine development in the context of HIV risk, and discuss implications and research needs for future HSV vaccine development.
Collapse
Affiliation(s)
- Joshua T Schiffer
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, WA, United States; Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, United States; University of Washington, Department of Medicine, Seattle, WA, United States.
| | - Sami L Gottlieb
- World Health Organization, Department of Reproductive Health and Research, Geneva, Switzerland
| |
Collapse
|
50
|
Ruebsamen-Schaeff H, Buschmann H. Different solid forms for optimizing route of administration of the herpes drug Pritelivir. MEDCHEMCOMM 2019; 10:1867-1870. [PMID: 32346467 DOI: 10.1039/c9md00233b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/05/2019] [Indexed: 11/21/2022]
Abstract
Pritelivir (AIC316, BAY 57-1293) was discovered as a highly potent drug against herpes simplex viruses with a novel mode of action, i.e. inhibition of the viral helicase-primase. A side by side comparison of the oral form against Valtrex™ in patients with genital herpes, showed superiority in phase II testing for Pritelivir. A number of different solid forms have been generated for additional, e.g. systemic, or topical applications.
Collapse
Affiliation(s)
| | - Helmut Buschmann
- AiCuris Anti-Infective Cures , Friedrich-Ebert-Strasse 475 , 42117 Wuppertal , Germany .
| |
Collapse
|