1
|
Birkmann A, Saunders R. Overview on the management of herpes simplex virus infections: Current therapies and future directions. Antiviral Res 2025; 237:106152. [PMID: 40154924 DOI: 10.1016/j.antiviral.2025.106152] [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: 01/10/2025] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
INTRODUCTION Herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) are highly prevalent, infecting approximately 64 % and 13 % of the world's population, respectively. Traditionally, HSV-1 has been associated with orofacial infections and HSV-2 with anogenital infections, but HSV-1 is increasingly the cause of genital infections. The clinical spectrum of HSV disease ranges from mild cold sores to severe conditions such as encephalitis or systemic infection, particularly in immunocompromised individuals and neonates. AREAS COVERED Here we summarize the natural history, epidemiology, manifestations, and treatment options for HSV infections. Current treatments, such as acyclovir, target viral DNA polymerase but have limited efficacy and are susceptible to resistance, especially in immunosuppressed populations. Rescue therapies such as foscarnet exhibit limiting toxicity. Vaccine development has been challenging, and a cure for HSV infection remains distant. Gene therapy is still in its early stages, while novel drugs such as helicase primase inhibitors (HPIs) are emerging as a promising alternative, showing high efficacy and the potential to overcome resistance. EXPERT OPINION HPIs represent a significant advance in HSV management. Their safety profile and novel mode of action may provide better viral suppression with a lower risk of resistance, offering hope for better control of the disease.
Collapse
Affiliation(s)
- Alexander Birkmann
- AiCuris Anti-infective Cures AG, Friedrich-Ebert-Str. 475, 42117, Wuppertal, Germany.
| | | |
Collapse
|
2
|
Casto AM, Song H, Xie H, Selke S, Roychoudhury P, Wu MC, Wald A, Greninger AL, Johnston C. Viral Genomic Variation and the Severity of Genital Herpes Simplex Virus-2 Infection as Quantified by Shedding Rate: A Viral Genome-Wide Association Study. J Infect Dis 2024; 230:1357-1366. [PMID: 38805234 PMCID: PMC11646587 DOI: 10.1093/infdis/jiae283] [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: 02/14/2024] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND The clinical severity of genital herpes simplex virus-2 (HSV-2) infection varies widely among infected persons with some experiencing frequent genital lesions while others are asymptomatic. The viral genital shedding rate is closely associated with, and has been established as, a surrogate marker of clinical severity. METHODS To assess the relationship between viral genetics and shedding, we assembled a set of 145 persons who had the severity of their genital herpes quantified through determination of their HSV genital shedding rate. An HSV-2 sample from each person was sequenced and biallelic variants among these genomes were identified. RESULTS We found no association between metrics of genome-wide variation in HSV-2 and shedding rate. A viral genome-wide association study identified the minor alleles of 3 individual unlinked variants as significantly associated with higher shedding rate (P < 8.4 × 10-5): C44973T (A512T), a nonsynonymous variant in UL22 (glycoprotein H); A74534G, a synonymous variant in UL36 (large tegument protein); and T119283C, an intergenic variant. We also found an association between the total number of minor alleles for the significant variants and shedding rate (P = 6.6 × 10-7). CONCLUSIONS These results add to a growing body of literature for HSV suggesting a connection between viral genetic variation and clinically important phenotypes of infection.
Collapse
Affiliation(s)
- Amanda M Casto
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Center, Seattle, Washington, USA
| | - Hoseung Song
- Division of Industrial and Systems Engineering, Graduate School of Data Science, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Hong Xie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Stacy Selke
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Pavitra Roychoudhury
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Center, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Michael C Wu
- Public Health Sciences Division, Fred Hutch Cancer Center, Seattle, Washington, USA
| | - Anna Wald
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Center, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Alexander L Greninger
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Center, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Christine Johnston
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Diseases Division, Fred Hutch Cancer Center, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
3
|
Parast L, Tian L, Cai T. Assessing heterogeneity in surrogacy using censored data. Stat Med 2024; 43:3184-3209. [PMID: 38812276 DOI: 10.1002/sim.10122] [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: 12/11/2023] [Revised: 04/22/2024] [Accepted: 05/10/2024] [Indexed: 05/31/2024]
Abstract
Determining whether a surrogate marker can be used to replace a primary outcome in a clinical study is complex. While many statistical methods have been developed to formally evaluate a surrogate marker, they generally do not provide a way to examine heterogeneity in the utility of a surrogate marker. Similar to treatment effect heterogeneity, where the effect of a treatment varies based on a patient characteristic, heterogeneity in surrogacy means that the strength or utility of the surrogate marker varies based on a patient characteristic. The few methods that have been recently developed to examine such heterogeneity cannot accommodate censored data. Studies with a censored outcome are typically the studies that could most benefit from a surrogate because the follow-up time is often long. In this paper, we develop a robust nonparametric approach to assess heterogeneity in the utility of a surrogate marker with respect to a baseline variable in a censored time-to-event outcome setting. In addition, we propose and evaluate a testing procedure to formally test for heterogeneity at a single time point or across multiple time points simultaneously. Finite sample performance of our estimation and testing procedure are examined in a simulation study. We use our proposed method to investigate the complex relationship between change in fasting plasma glucose, diabetes, and sex hormones using data from the diabetes prevention program study.
Collapse
Affiliation(s)
- Layla Parast
- Department of Statistics and Data Sciences, The University of Texas at Austin, Austin, Texas
| | - Lu Tian
- Department of Biomedical Data Science, Stanford University, Stanford, California
| | - Tianxi Cai
- Department of Biostatistics, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
4
|
Johnston C, Scheele S, Bachmann L, Boily MC, Chaiyakunapruk N, Deal C, Delany-Moretlwe S, Lee S, Looker K, Marshall C, Mello MB, Ndowa F, Gottlieb S. Vaccine value profile for herpes simplex virus. Vaccine 2024; 42:S82-S100. [PMID: 39003018 DOI: 10.1016/j.vaccine.2024.01.044] [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: 08/01/2023] [Revised: 10/10/2023] [Accepted: 01/15/2024] [Indexed: 07/15/2024]
Abstract
Herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) are chronic, highly prevalent viral infections that cause significant morbidity around the world. HSV-2 is sexually transmitted and is the leading cause of genital ulcer disease (GUD). It also increases the risk of HIV acquisition, fueling the HIV epidemic. HSV-1 is typically acquired in childhood through nonsexual contact and contributes to oral and ocular disease, but it can also be sexually transmitted to cause GUD. Both HSV-1 and HSV-2 cause neonatal herpes and neurologic disease. Given the ubiquitous nature of HSV-1 and HSV-2 infections and the limited existing prevention and control measures, vaccination would be the most efficient strategy to reduce the global burden of morbidity related to HSV infection. Vaccine strategies include prophylactic vaccination, which would prevent infection among susceptible persons and would likely be given to adolescents, and therapeutic vaccinations, which would be given to people with symptomatic genital HSV-2 infection. This document discusses the vaccine value profile of both types of vaccines. This 'Vaccine Value Profile' (VVP) for HSV is intended to provide a high-level, holistic assessment of the information and data that are currently available to inform the potential public health, economic and societal value of pipeline vaccines and vaccine-like products. This VVP was developed by subject matter experts from academia, non-profit organizations, government agencies and multi-lateral organizations. All contributors have extensive expertise on various elements of the HSV VVP and collectively aimed to identify current research and knowledge gaps. The VVP was developed using only existing and publicly available information.
Collapse
Affiliation(s)
- Christine Johnston
- Division of Allergy & Infectious Diseases, University of Washington, Seattle, WA, USA.
| | - Suzanne Scheele
- Center for Vaccine Introduction and Access, PATH, Washington, DC, USA
| | - Laura Bachmann
- Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Marie-Claude Boily
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, UK
| | - Nathorn Chaiyakunapruk
- Department of Pharmacotherapy, University of Utah College of Pharmacy, Salt Lake City, UT, USA
| | - Carolyn Deal
- Enteric and Sexually Transmitted Diseases Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | | | - Shaun Lee
- Monash University Malaysia, Subang, Malaysia
| | - Katharine Looker
- Population Health Sciences, Bristol Medical School, University of Bristol, UK
| | - Caroline Marshall
- Department of Immunizations, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| | - Maeve B Mello
- Global HIV, Hepatitis and STI Programmes, World Health Organization, Geneva, Switzerland
| | | | - Sami Gottlieb
- Department of Sexual and Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| |
Collapse
|
5
|
Parast L, Tian L, Cai T, Palaniappan L. Statistical Methods to Evaluate Surrogate Markers. Med Care 2024; 62:102-108. [PMID: 38079232 PMCID: PMC10842261 DOI: 10.1097/mlr.0000000000001956] [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] [Indexed: 01/12/2024]
Abstract
BACKGROUND There is tremendous interest in evaluating surrogate markers given their potential to decrease study time, costs, and patient burden. OBJECTIVES The purpose of this statistical workshop article is to describe and illustrate how to evaluate a surrogate marker of interest using the proportion of treatment effect (PTE) explained as a measure of the quality of the surrogate marker for: (1) a setting with a general fully observed primary outcome (eg, biopsy score); and (2) a setting with a time-to-event primary outcome which may be censored due to study termination or early drop out (eg, time to diabetes). METHODS The methods are motivated by 2 randomized trials, one among children with nonalcoholic fatty liver disease where the primary outcome was a change in biopsy score (general outcome) and another study among adults at high risk for Type 2 diabetes where the primary outcome was time to diabetes (time-to-event outcome). The methods are illustrated using the Rsurrogate package with a detailed R code provided. RESULTS In the biopsy score outcome setting, the estimated PTE of the examined surrogate marker was 0.182 (95% confidence interval [CI]: 0.121, 0.240), that is, the surrogate explained only 18.2% of the treatment effect on the biopsy score. In the diabetes setting, the estimated PTE of the surrogate marker was 0.596 (95% CI: 0.404, 0.760), that is, the surrogate explained 59.6% of the treatment effect on diabetes incidence. CONCLUSIONS This statistical workshop provides tools that will support future researchers in the evaluation of surrogate markers.
Collapse
Affiliation(s)
- Layla Parast
- Department of Statistics and Data Science, The University of Texas at Austin, Austin, TX, USA
| | - Lu Tian
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Tianxi Cai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Latha Palaniappan
- Department of Medicine, Stanford University, School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
6
|
Parast L, Tian L, Cai T, Palaniappan LP. Can earlier biomarker measurements explain a treatment effect on diabetes incidence? A robust comparison of five surrogate markers. BMJ Open Diabetes Res Care 2023; 11:e003585. [PMID: 37907279 PMCID: PMC10619035 DOI: 10.1136/bmjdrc-2023-003585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 10/07/2023] [Indexed: 11/02/2023] Open
Abstract
INTRODUCTION We measured and compared five individual surrogate markers-change from baseline to 1 year after randomization in hemoglobin A1c (HbA1c), fasting glucose, 2-hour postchallenge glucose, triglyceride-glucose index (TyG) index, and homeostatic model assessment of insulin resistance (HOMA-IR)-in terms of their ability to explain a treatment effect on reducing the risk of type 2 diabetes mellitus at 2, 3, and 4 years after treatment initiation. RESEARCH DESIGN AND METHODS Study participants were from the Diabetes Prevention Program study, randomly assigned to either a lifestyle intervention (n=1023) or placebo (n=1030). The surrogate markers were measured at baseline and 1 year, and diabetes incidence was examined at 2, 3, and 4 years postrandomization. Surrogacy was evaluated using a robust model-free estimate of the proportion of treatment effect explained (PTE) by the surrogate marker. RESULTS Across all time points, change in fasting glucose and HOMA-IR explained higher proportions of the treatment effect than 2-hour glucose, TyG index, or HbA1c. For example, at 2 years, glucose explained the highest (80.1%) proportion of the treatment effect, followed by HOMA-IR (77.7%), 2-hour glucose (76.2%), and HbA1c (74.6%); the TyG index explained the smallest (70.3%) proportion. CONCLUSIONS These data suggest that, of the five examined surrogate markers, glucose and HOMA-IR were the superior surrogate markers in terms of PTE, compared with 2-hour glucose, HbA1c, and TyG index.
Collapse
Affiliation(s)
- Layla Parast
- The University of Texas at Austin, Austin, Texas, USA
| | - Lu Tian
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
| | - Tianxi Cai
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, Massachusetts, USA
| | - Latha P Palaniappan
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
7
|
Wang X, Parast L, Han L, Tian L, Cai T. Robust approach to combining multiple markers to improve surrogacy. Biometrics 2023; 79:788-798. [PMID: 35426444 PMCID: PMC10347081 DOI: 10.1111/biom.13677] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 03/28/2022] [Indexed: 12/01/2022]
Abstract
Identifying effective and valid surrogate markers to make inference about a treatment effect on long-term outcomes is an important step in improving the efficiency of clinical trials. Replacing a long-term outcome with short-term and/or cheaper surrogate markers can potentially shorten study duration and reduce trial costs. There is sizable statistical literature on methods to quantify the effectiveness of a single surrogate marker. Both parametric and nonparametric approaches have been well developed for different outcome types. However, when there are multiple markers available, methods for combining markers to construct a composite marker with improved surrogacy remain limited. In this paper, building on top of the optimal transformation framework of Wang et al. (2020), we propose a novel calibrated model fusion approach to optimally combine multiple markers to improve surrogacy. Specifically, we obtain two initial estimates of optimal composite scores of the markers based on two sets of models with one set approximating the underlying data distribution and the other directly approximating the optimal transformation function. We then estimate an optimal calibrated combination of the two estimated scores which ensures both validity of the final combined score and optimality with respect to the proportion of treatment effect explained by the final combined score. This approach is unique in that it identifies an optimal combination of the multiple surrogates without strictly relying on parametric assumptions while borrowing modeling strategies to avoid fully nonparametric estimation which is subject to the curse of dimensionality. Our identified optimal transformation can also be used to directly quantify the surrogacy of this identified combined score. Theoretical properties of the proposed estimators are derived, and the finite sample performance of the proposed method is evaluated through simulation studies. We further illustrate the proposed method using data from the Diabetes Prevention Program study.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Biostatistics, Harvard University, Boston, Massachusetts, USA
| | - Layla Parast
- Department of Statistics and Data Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - Larry Han
- Department of Biostatistics, Harvard University, Boston, Massachusetts, USA
| | - Lu Tian
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
| | - Tianxi Cai
- Department of Biostatistics, Harvard University, Boston, Massachusetts, USA
- Department of Biomedical Informatics, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Parast L, Cai T, Tian L. Testing for heterogeneity in the utility of a surrogate marker. Biometrics 2023; 79:799-810. [PMID: 34874550 PMCID: PMC9170832 DOI: 10.1111/biom.13600] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/12/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022]
Abstract
In studies that require long-term and/or costly follow-up of participants to evaluate a treatment, there is often interest in identifying and using a surrogate marker to evaluate the treatment effect. While several statistical methods have been proposed to evaluate potential surrogate markers, available methods generally do not account for or address the potential for a surrogate to vary in utility or strength by patient characteristics. Previous work examining surrogate markers has indicated that there may be such heterogeneity, that is, that a surrogate marker may be useful (with respect to capturing the treatment effect on the primary outcome) for some subgroups, but not for others. This heterogeneity is important to understand, particularly if the surrogate is to be used in a future trial to replace the primary outcome. In this paper, we propose an approach and estimation procedures to measure the surrogate strength as a function of a baseline covariate W and thus examine potential heterogeneity in the utility of the surrogate marker with respect to W. Within a potential outcome framework, we quantify the surrogate strength/utility using the proportion of treatment effect on the primary outcome that is explained by the treatment effect on the surrogate. We propose testing procedures to test for evidence of heterogeneity, examine finite sample performance of these methods via simulation, and illustrate the methods using AIDS clinical trial data.
Collapse
Affiliation(s)
- Layla Parast
- Statistics Group, RAND Corporation, Santa Monica, California, USA
| | - Tianxi Cai
- Department of Biostatistics, Harvard University, Boston, Massachusetts, USA
| | - Lu Tian
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
| |
Collapse
|
9
|
Laing KJ, Ouwendijk WJD, Campbell VL, McClurkan CL, Mortazavi S, Elder Waters M, Krist MP, Tu R, Nguyen N, Basu K, Miao C, Schmid DS, Johnston C, Verjans GMGM, Koelle DM. Selective retention of virus-specific tissue-resident T cells in healed skin after recovery from herpes zoster. Nat Commun 2022; 13:6957. [PMID: 36376285 PMCID: PMC9663441 DOI: 10.1038/s41467-022-34698-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022] Open
Abstract
Herpes zoster is a localized skin infection caused by reactivation of latent varicella-zoster virus. Tissue-resident T cells likely control skin infections. Zoster provides a unique opportunity to determine if focal reinfection of human skin boosts local or disseminated antigen-specific tissue-resident T cells. Here, we show virus-specific T cells are retained over one year in serial samples of rash site and contralateral unaffected skin of individuals recovered from zoster. Consistent with zoster resolution, viral DNA is largely undetectable on skin from day 90 and virus-specific B and T cells decline in blood. In skin, there is selective infiltration and long-term persistence of varicella-zoster virus-specific T cells in the rash site relative to the contralateral site. The skin T cell infiltrates express the canonical tissue-resident T cell markers CD69 and CD103. These findings show that zoster promotes spatially-restricted long-term retention of antigen-specific tissue-resident T cells in previously infected skin.
Collapse
Affiliation(s)
- Kerry J Laing
- Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Werner J D Ouwendijk
- HerpeslabNL of the Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Shahin Mortazavi
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Maxwell P Krist
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Richard Tu
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Nhi Nguyen
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Krithi Basu
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Congrong Miao
- Centers for Disease Control and Prevention, Division of Viral Diseases, Atlanta, GA, USA
| | - D Scott Schmid
- Centers for Disease Control and Prevention, Division of Viral Diseases, Atlanta, GA, USA
| | - Christine Johnston
- Department of Medicine, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Georges M G M Verjans
- HerpeslabNL of the Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - David M Koelle
- Department of Medicine, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Translational Research, Benaroya Research Institute, Seattle, WA, USA
| |
Collapse
|
10
|
Gompels UA, Bravo FJ, Briggs S, Ameri S, Cardin RD, Bernstein DI. Immunisation Using Novel DNA Vaccine Encoding Virus Membrane Fusion Complex and Chemokine Genes Shows High Protection from HSV-2. Viruses 2022; 14:v14112317. [PMID: 36366414 PMCID: PMC9698128 DOI: 10.3390/v14112317] [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: 09/30/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022] Open
Abstract
Herpes simplex virus 1 and 2 infections cause high unmet disease burdens worldwide. Mainly HSV-2 causes persistent sexually transmitted disease, fatal neonatal disease and increased transmission of HIV/AIDS. Thus, there is an urgent requirement to develop effective vaccines. We developed nucleic acid vaccines encoding a novel virus entry complex stabilising cell membrane fusion, 'virus-like membranes', VLM. Two dose intramuscular immunisations using DNA expression plasmids in a guinea pig model gave 100% protection against acute disease and significantly reduced virus replication after virus intravaginal challenge. There was also reduced establishment of latency within the dorsal root ganglia and spinal cord, but recurrent disease and recurrent virus shedding remained. To increase cellular immunity and protect against recurrent disease, cDNA encoding an inhibitor of chemokine receptors on T regulatory cells was added and compared to chemokine CCL5 effects. Immunisation including this novel human chemokine gene, newly defined splice variant from an endogenous virus genome, 'virokine immune therapeutic', VIT, protected most guinea pigs from recurrent disease and reduced recurrent virus shedding distinct from a gD protein vaccine similar to that previously evaluated in clinical trials. All DNA vaccines induced significant neutralising antibodies and warrant evaluation for new therapeutic treatments.
Collapse
Affiliation(s)
- Ursula A. Gompels
- Virothera, Milner Therapeutics Institute, Cambridge Biomedical Campus, Cambridge CB4 0WS, UK
- Correspondence:
| | - Fernando J. Bravo
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Sean Briggs
- Virothera, Milner Therapeutics Institute, Cambridge Biomedical Campus, Cambridge CB4 0WS, UK
| | - Shima Ameri
- Virothera, Milner Therapeutics Institute, Cambridge Biomedical Campus, Cambridge CB4 0WS, UK
| | - Rhonda D. Cardin
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - David I. Bernstein
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
11
|
Waghmare A, Krantz EM, Baral S, Vasquez E, Loeffelholz T, Chung EL, Pandey U, Kuypers J, Duke ER, Jerome KR, Greninger AL, Reeves DB, Hladik F, Cardozo-Ojeda EF, Boeckh M, Schiffer JT. Reliability of Self-Sampling for Accurate Assessment of Respiratory Virus Viral and Immunologic Kinetics. J Infect Dis 2022; 226:278-286. [PMID: 32710762 PMCID: PMC7454707 DOI: 10.1093/infdis/jiaa451] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic demonstrates the need for accurate and convenient approaches to diagnose and therapeutically monitor respiratory viral infections. We demonstrated that self-sampling with mid-nasal foam swabs is well-tolerated and provides quantitative viral output concordant with flocked swabs. Using longitudinal home-based self-sampling, we demonstrate that nasal cytokine levels correlate and cluster according to immune cell of origin. Periods of stable viral loads are followed by rapid elimination, which could be coupled with cytokine expansion and contraction. Nasal foam swab self-sampling at home provides a precise, mechanistic readout of respiratory virus shedding and local immune responses.
Collapse
Affiliation(s)
- Alpana Waghmare
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Center for Clinical and Translational Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Elizabeth M Krantz
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Subhasish Baral
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Emma Vasquez
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Tillie Loeffelholz
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - E Lisa Chung
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Urvashi Pandey
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Jane Kuypers
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Elizabeth R Duke
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Keith R Jerome
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Alexander L Greninger
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Daniel B Reeves
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Florian Hladik
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - E Fabian Cardozo-Ojeda
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Michael Boeckh
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Joshua T Schiffer
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
12
|
Pan X, Zhang Y, Zhao Y, Yao S, Guan C, Wang L, Chen L. Inhibitory activity and mechanism of silver nanoparticles against herpes simplex virus type 1. Arch Virol 2022; 167:1619-1636. [PMID: 35648293 DOI: 10.1007/s00705-022-05467-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 04/26/2022] [Indexed: 12/01/2022]
Abstract
Herpes simplex virus type 1 (HSV-1) is a common pathogen that infects 50-90% of the world's population and causes a variety of diseases, some of which can be life-threatening. Silver nanoparticles (AgNPs) have been shown to have broad-spectrum antiviral activity. In this study, we investigated the activity of AgNPs against HSV-1 and found that AgNPs effectively inhibited plaque formation and HSV-1 progeny production, reduced the genomic load, and interfered with HSV-1 mRNA expression and protein synthesis. Transmission electron microscopy showed that AgNPs interacted with HSV-1 and altered the shape of the viral particles. Furthermore, AgNPs affected the entry of HSV-1 into cells as well as their release and cell-to-cell spread. AgNPs were also found to downregulate the expression of pro-inflammatory cytokines upon HSV-1 infection. Combined treatment with AgNPs and acyclovir (ACV) confirmed that AgNPs significantly enhanced the inhibitory effect of ACV against HSV-1. Our findings may contribute to an understanding of the mechanism of the antiviral effect of AgNPs against HSV-1 and help to provide a theoretical basis for their clinical application.
Collapse
Affiliation(s)
- Xuanhe Pan
- Department of Clinical Laboratory, Liuzhou People's Hospital, Liuzhou, Guangxi, China
| | - Yapeng Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yiming Zhao
- Department of Medical Microbiology, School of Basic Medical Sciences, Central South University, No. 172, Tongzipo road, Yuelu District, Changsha, 410013, Hunan, China
| | - Siqi Yao
- Department of Medical Microbiology, School of Basic Medical Sciences, Central South University, No. 172, Tongzipo road, Yuelu District, Changsha, 410013, Hunan, China
| | - Chaxiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Linqian Wang
- Department of Clinical Laboratory, the Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China.
| | - Liyu Chen
- Department of Medical Microbiology, School of Basic Medical Sciences, Central South University, No. 172, Tongzipo road, Yuelu District, Changsha, 410013, Hunan, China.
| |
Collapse
|
13
|
Koelle DM, Dong L, Jing L, Laing KJ, Zhu J, Jin L, Selke S, Wald A, Varon D, Huang ML, Johnston C, Corey L, Posavad CM. HSV-2-Specific Human Female Reproductive Tract Tissue Resident Memory T Cells Recognize Diverse HSV Antigens. Front Immunol 2022; 13:867962. [PMID: 35432373 PMCID: PMC9009524 DOI: 10.3389/fimmu.2022.867962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/07/2022] [Indexed: 01/05/2023] Open
Abstract
Antigen-specific TRM persist and protect against skin or female reproductive tract (FRT) HSV infection. As the pathogenesis of HSV differs between humans and model organisms, we focus on humans with well-characterized recurrent genital HSV-2 infection. Human CD8+ TRM persisting at sites of healed human HSV-2 lesions have an activated phenotype but it is unclear if TRM can be cultivated in vitro. We recovered HSV-specific TRM from genital skin and ectocervix biopsies, obtained after recovery from recurrent genital HSV-2, using ex vivo activation by viral antigen. Up to several percent of local T cells were HSV-reactive ex vivo. CD4 and CD8 T cell lines were up to 50% HSV-2-specific after sorting-based enrichment. CD8 TRM displayed HLA-restricted reactivity to specific HSV-2 peptides with high functional avidities. Reactivity to defined peptides persisted locally over several month and was quite subject-specific. CD4 TRM derived from biopsies, and from an extended set of cervical cytobrush specimens, also recognized diverse HSV-2 antigens and peptides. Overall we found that HSV-2-specific TRM are abundant in the FRT between episodes of recurrent genital herpes and maintain competency for expansion. Mucosal sites are accessible for clinical monitoring during immune interventions such as therapeutic vaccination.
Collapse
Affiliation(s)
- David M. Koelle
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Translational Research, Benaroya Research Institute, Seattle, WA, United States
| | - Lichun Dong
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Lichen Jing
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Kerry J. Laing
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Jia Zhu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Lei Jin
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Stacy Selke
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Anna Wald
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Dana Varon
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Meei-Li Huang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Christine Johnston
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Lawrence Corey
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Christine M. Posavad
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| |
Collapse
|
14
|
Zhou RR, Zhao SD, Parast L. Estimation of the proportion of treatment effect explained by a high-dimensional surrogate. Stat Med 2022; 41:2227-2246. [PMID: 35189671 DOI: 10.1002/sim.9352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/23/2021] [Accepted: 01/27/2022] [Indexed: 11/07/2022]
Abstract
Clinical studies examining the effectiveness of a treatment with respect to some primary outcome often require long-term follow-up of patients and/or costly or burdensome measurements of the primary outcome of interest. Identifying a surrogate marker for the primary outcome of interest may allow one to evaluate a treatment effect with less follow-up time, less cost, or less burden. While much clinical and statistical work has focused on identifying and validating surrogate markers, available approaches tend to focus on settings in which only a single surrogate marker is of interest. Limited work has been done to accommodate the high-dimensional surrogate marker setting where the number of potential surrogates is greater than the sample size. In this article, we develop methods to estimate the proportion of treatment effect explained by high-dimensional surrogates. We study the asymptotic properties of our proposed estimator, propose inference procedures, and examine finite sample performance via a simulation study. We illustrate our proposed methods using data from a randomized study comparing a novel whey-based oral nutrition supplement with a standard supplement with respect to change in body fat percentage over 12 weeks, where the surrogate markers of interest are gene expression probesets.
Collapse
Affiliation(s)
| | - Sihai Dave Zhao
- Department of Statistics, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Layla Parast
- Department of Statistics and Data Sciences, University of Texas at Austin, Austin, USA
| |
Collapse
|
15
|
White JM, Schiffer JT, Bender Ignacio RA, Xu S, Kainov D, Ianevski A, Aittokallio T, Frieman M, Olinger GG, Polyak SJ. Drug Combinations as a First Line of Defense against Coronaviruses and Other Emerging Viruses. mBio 2021; 12:e0334721. [PMID: 34933447 PMCID: PMC8689562 DOI: 10.1128/mbio.03347-21] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The world was unprepared for coronavirus disease 2019 (COVID-19) and remains ill-equipped for future pandemics. While unprecedented strides have been made developing vaccines and treatments for COVID-19, there remains a need for highly effective and widely available regimens for ambulatory use for novel coronaviruses and other viral pathogens. We posit that a priority is to develop pan-family drug cocktails to enhance potency, limit toxicity, and avoid drug resistance. We urge cocktail development for all viruses with pandemic potential both in the short term (<1 to 2 years) and longer term with pairs of drugs in advanced clinical testing or repurposed agents approved for other indications. While significant efforts were launched against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), in vitro and in the clinic, many studies employed solo drugs and had disappointing results. Here, we review drug combination studies against SARS-CoV-2 and other viruses and introduce a model-driven approach to assess drug pairs with the highest likelihood of clinical efficacy. Where component agents lack sufficient potency, we advocate for synergistic combinations to achieve therapeutic levels. We also discuss issues that stymied therapeutic progress against COVID-19, including testing of agents with low likelihood of efficacy late in clinical disease and lack of focus on developing virologic surrogate endpoints. There is a need to expedite efficient clinical trials testing drug combinations that could be taken at home by recently infected individuals and exposed contacts as early as possible during the next pandemic, whether caused by a coronavirus or another viral pathogen. The approach herein represents a proactive plan for global viral pandemic preparedness.
Collapse
Affiliation(s)
- Judith M. White
- University of Virginia, Department of Cell Biology, Charlottesville, Virginia, USA
- University of Virginia, Department of Microbiology, Charlottesville, Virginia, USA
| | - Joshua T. Schiffer
- University of Washington, Division of Allergy and Infectious Diseases, Seattle, Washington, USA
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Rachel A. Bender Ignacio
- University of Washington, Division of Allergy and Infectious Diseases, Seattle, Washington, USA
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Shuang Xu
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Denis Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Institute of Technology, University of Tartu, Tartu, Estonia
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Aleksandr Ianevski
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
- Oslo Centre for Biostatistics and Epidemiology (OCBE), University of Oslo, Oslo, Norway
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Matthew Frieman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Stephen J. Polyak
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
16
|
Hilario GM, Sulczewski FB, Liszbinski R, Mello LD, Hagen G, Fazolo T, Neto J, Dallegrave E, Romão P, Aguirre T, Rodrigues Junior LC. Development and immunobiological evaluation of nanoparticles containing an immunodominant epitope of herpes simplex virus. IET Nanobiotechnol 2021; 15:532-544. [PMID: 34694744 PMCID: PMC8675790 DOI: 10.1049/nbt2.12043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 11/19/2022] Open
Abstract
Herpes simplex virus (HSV) 1 and 2 are viruses that infect individuals worldwide and for which there is no cure or vaccine available. The protective response against herpes is mostly mediated by CD8 T lymphocytes that respond to the immunodominant SSIEFARL epitope. However, there are some obstacles concerning the use of free SSIEFARL for vaccine or immunotherapy. The aim of this study was to evaluate the feasibility of nanoencapsulation of SSIEFARL and its immunostimulatory properties. Nano/SSIEFARL was produced by interfacial polymerization in methylmetacrylate, and the physico-chemical properties, morphology and immunobiological parameters were evaluated. To evaluate the ex vivo capacity of Nano/SSIEFARL, we used splenocytes from HSV-1-infected mice to enhance the frequency of SSIEFARL-specific CD8 T lymphocytes. The results indicate that Nano/SSIEFARL has a spherical shape, an average diameter of 352 ± 22 nm, the PDI was 0.361 ± 0.009 and is negatively charged (-26.30 ± 35). The stability at 4°C was 28 days. Also, Nano/SSIEFARL is not toxic for cells at low concentrations in vitro and it is taken up by JAWS II dendritic cells. No histopathological changes were observed in kidneys, liver and lymph nodes of animals treated with Nano/SSIEFARL. Nan/SSIEFARL increased the production of IL-1β, TNF-α and IL-12 by the dendritic cells. Finally, Nano/SSIEFARL expanded the frequency of SSIEFARL-specific CD8+T lymphocytes at the same rate as free SSIEFARL. In conclusion all data together indicate that SSIEFARL is suitable for nanoencapsulation, and the system produced presents some immunoadjuvant properties that can be used to improve the immune response against herpes.
Collapse
Affiliation(s)
- Gabriel M Hilario
- Laboratorio de Imunovirologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| | - Fernando B Sulczewski
- Laboratorio de Imunovirologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| | - Raquel Liszbinski
- Laboratorio de Imunovirologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| | - Larissa D Mello
- Laboratório de Nanotecnologia, Universidade Franciscana, Brazil
| | - Gustavo Hagen
- Laboratorio de Imunovirologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| | - Tiago Fazolo
- Laboratório de Imunologia Celular e Molecular, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| | - Jayme Neto
- Laboratorio de Imunovirologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| | - Eliane Dallegrave
- Laboratório de Pesquisa em Toxicologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| | - Pedro Romão
- Laboratório de Imunologia Celular e Molecular, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| | - Tanira Aguirre
- Laboratório de Imunoterapia, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| | - Luiz C Rodrigues Junior
- Laboratorio de Imunovirologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| |
Collapse
|
17
|
Stinn T, Kuntz S, Varon D, Huang ML, Selke S, Njikan S, Ford ES, Dragavon J, Coombs RW, Johnston C, Bull ME. Subclinical Genital Herpes Shedding in HIV/Herpes Simplex Virus 2-Coinfected Women during Antiretroviral Therapy Is Associated with an Increase in HIV Tissue Reservoirs and Potentially Promotes HIV Evolution. J Virol 2020; 95:e01606-20. [PMID: 33028713 PMCID: PMC7737750 DOI: 10.1128/jvi.01606-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/17/2020] [Indexed: 01/18/2023] Open
Abstract
Antigen (Ag)-specific immune responses to chronic infections, such as herpes simplex virus type 2 (HSV-2) in HIV/HSV-coinfected persons, may sustain HIV tissue reservoirs by promoting T-cell proliferation but are poorly studied in women on antiretroviral therapy (ART). Mixed anogenital swabs and cervical secretions were self-collected by nine HIV/HSV-2-coinfected women during ART for 28 days to establish subclinical HSV DNA shedding rates and detection of HIV RNA by real-time PCR. Typical herpes lesion site biopsy (TLSB) and cervical biopsy specimens were collected at the end of the daily sampling period. Nucleic acids (NA) isolated from biopsy specimens had HIV quantified and HIV envC2-V5 single-genome amplification (SGA) and T-cell receptor (TCR) repertoires assessed. Women had a median CD4 count of 537 cells/μl (IQR: 483 to 741) at enrollment and HIV plasma viral loads of <40 copies/ml. HSV DNA was detected on 12% of days (IQR: 2 to 25%) from anogenital specimens. Frequent subclinical HSV DNA shedding was associated with increased HIV DNA tissue concentrations and increased divergence from the most recent common ancestor (MRCA), an indicator of HIV replication. Distinct predominant TCR clones were detected in cervical and TLSB specimens in a woman with frequent HSV DNA shedding, with mixing of minor variants between her tissues. In contrast, more limited TCR repertoire mixing was observed in two women with less frequent subclinical HSV DNA shedding. Subclinical HSV shedding in HIV/HSV-coinfected women during ART may sustain HIV tissue reservoirs via Ag exposure or HIV replication. This study provides evidence supporting further study of interventions targeting suppression of Ag-specific immune responses as a component of HIV cure strategies.IMPORTANCE Persons with HIV infection are frequently coinfected with chronic herpesviruses, which periodically replicate and produce viable herpes virions, particularly in anogenital and cervical tissues. Persistent protein expression results in proliferation of CD8+ and CD4+ T cells, and the latter could potentially expand and sustain HIV tissue reservoirs. We found HSV genital shedding rates were positively correlated with HIV DNA concentrations and HIV divergence from ancestral sequences in tissues. Our work suggests that immune responses to common coinfections, such as herpesviruses, may sustain HIV tissue reservoirs during suppressive ART, suggesting future cure strategies should study interventions to suppress replication or reactivation of chronic herpes infections.
Collapse
Affiliation(s)
- Tajanna Stinn
- Seattle Children's Research Institute, Seattle, Washington, USA
| | - Steve Kuntz
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Dana Varon
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Meei-Li Huang
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Stacy Selke
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Samuel Njikan
- Seattle Children's Research Institute, Seattle, Washington, USA
| | - Emily S Ford
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Joan Dragavon
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Robert W Coombs
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Christine Johnston
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Marta E Bull
- Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
18
|
Parast L, Garcia TP, Prentice RL, Carroll RJ. Robust methods to correct for measurement error when evaluating a surrogate marker. Biometrics 2020; 78:9-23. [PMID: 33021738 DOI: 10.1111/biom.13386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/16/2020] [Accepted: 09/30/2020] [Indexed: 11/27/2022]
Abstract
The identification of valid surrogate markers of disease or disease progression has the potential to decrease the length and costs of future studies. Most available methods that assess the value of a surrogate marker ignore the fact that surrogates are often measured with error. Failing to adjust for measurement error can erroneously identify a useful surrogate marker as not useful or vice versa. We investigate and propose robust methods to correct for the effect of measurement error when evaluating a surrogate marker using multiple estimators developed for parametric and nonparametric estimates of the proportion of treatment effect explained by the surrogate marker. In addition, we quantify the attenuation bias induced by measurement error and develop inference procedures to allow for variance and confidence interval estimation. Through a simulation study, we show that our proposed estimators correct for measurement error in the surrogate marker and that our inference procedures perform well in finite samples. We illustrate these methods by examining a potential surrogate marker that is measured with error, hemoglobin A1c, using data from the Diabetes Prevention Program clinical trial.
Collapse
Affiliation(s)
- Layla Parast
- RAND Corporation, Statistics Group, Santa Monica, California
| | - Tanya P Garcia
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ross L Prentice
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Raymond J Carroll
- Department of Statistics, Texas A&M University, College Station, Texas.,School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway, NSW, Australia
| |
Collapse
|
19
|
Waghmare A, Krantz EM, Baral S, Vasquez E, Loeffelholz T, Chung EL, Pandey U, Kuypers J, Duke ER, Jerome KR, Greninger AL, Reeves DB, Hladik F, Cardozo-Ojeda EF, Boeckh M, Schiffer JT. Reliability of self-sampling for accurate assessment of respiratory virus viral and immunologic kinetics. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.04.03.20051706. [PMID: 32511581 PMCID: PMC7276008 DOI: 10.1101/2020.04.03.20051706] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
The SARS-CoV-2 pandemic demonstrates the need for accurate and convenient approaches to diagnose and therapeutically monitor respiratory viral infections. We demonstrated that self-sampling with foam swabs at home is well-tolerated and provides quantitative viral output concordant with flocked swabs. Nasal cytokine levels correlate and cluster according to immune cell of origin. Periods of stable viral loads are followed by rapid elimination, which could be coupled with cytokine expansion and contraction using mathematical models. Nasal foam swab self-sampling at home provides a precise, mechanistic readout of respiratory virus shedding and local immune responses.
Collapse
Affiliation(s)
- Alpana Waghmare
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
- Department of Pediatrics, University of Washington
- Center for Clinical and Translational Research, Seattle Children’s Research Institute
| | - Elizabeth M. Krantz
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
| | - Subhasish Baral
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
| | - Emma Vasquez
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
| | - Tillie Loeffelholz
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
| | - E. Lisa Chung
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
| | - Urvashi Pandey
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
- Department of Obstetrics and Gynecology, University of Washington
| | - Jane Kuypers
- Department of Laboratory Medicine, University of Washington
| | - Elizabeth R Duke
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
- Department of Medicine, University of Washington
| | - Keith R. Jerome
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
- Department of Laboratory Medicine, University of Washington
| | | | - Daniel B. Reeves
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
| | - Florian Hladik
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
- Department of Obstetrics and Gynecology, University of Washington
- Department of Medicine, University of Washington
| | | | - Michael Boeckh
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
- Department of Medicine, University of Washington
- Clinical Research Division, Fred Hutchinson Cancer Research Center
| | - Joshua T. Schiffer
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center
- Department of Medicine, University of Washington
- Clinical Research Division, Fred Hutchinson Cancer Research Center
| |
Collapse
|
20
|
Szöllősi A, Raffai T, Bogdanov A, Endrész V, Párducz L, Somogyvári F, Janovák L, Burián K, Virok DP. Correlation between detergent activity and anti-herpes simplex virus-2 activity of commercially available vaginal gels. BMC Res Notes 2020; 13:52. [PMID: 32005126 PMCID: PMC6995179 DOI: 10.1186/s13104-020-4918-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 01/23/2020] [Indexed: 09/03/2023] Open
Abstract
OBJECTIVE Herpes simplex virus-2 (HSV-2) infections are almost exclusively sexually transmitted. The presence of vaginal gels during sexual activity may have a significant positive or negative impact on viral transmission. Therefore we investigated three off-the-shelf vaginal lubricants and one pH restoring gel to evaluate their impact on HSV-2 replication. RESULTS HeLa cells were infected with untreated virions and virions incubated with the particular gels. The accumulation of viral genomes was monitored by quantitative PCR (qPCR) method at 24 h post infection. Two of the tested gels had no significant effect on HSV-2 replication at the maximum applied concentration, while two had a strong inhibitory effect (~ 98% reduction of replication). The replication inhibitory effect was observed at various multiplicity of infection (MOI 0.4-6.4) and the two inhibitory gels were also capable of inhibiting the HSV-2 induced cytopathic effect on HeLa cells. The surface tension decreasing activity-an indication of detergent activity-was strongly correlated with the anti-HSV-2 activity of the gels (R2: 0.88). Our results indicate that off-the-shelf vaginal gels have a markedly different anti-HSV-2 activity that may influence HSV-2 transmission.
Collapse
Affiliation(s)
- Andrea Szöllősi
- Department of Health and Social Sciences, Gál Ferenc College, Szent István st. 17-19, Gyula, 5700, Hungary
| | - Tímea Raffai
- Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm sqr. 10, Szeged, 6720, Hungary
| | - Anita Bogdanov
- Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm sqr. 10, Szeged, 6720, Hungary
| | - Valéria Endrész
- Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm sqr. 10, Szeged, 6720, Hungary
| | - László Párducz
- Department of Health and Social Sciences, Gál Ferenc College, Szent István st. 17-19, Gyula, 5700, Hungary.,Pándy Kálmán County Hospital, Semmelweis st. 1, Gyula, 5700, Hungary
| | - Ferenc Somogyvári
- Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm sqr. 10, Szeged, 6720, Hungary
| | - László Janovák
- Interdisciplinary Excellence Centre, Department of Physical Chemistry and Materials Science, University of Szeged, Rerrich Béla sqr. 1, Szeged, 6720, Hungary
| | - Katalin Burián
- Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm sqr. 10, Szeged, 6720, Hungary
| | - Dezső P Virok
- Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm sqr. 10, Szeged, 6720, Hungary.
| |
Collapse
|
21
|
Chandra J, Woo WP, Dutton JL, Xu Y, Li B, Kinrade S, Druce J, Finlayson N, Griffin P, Laing KJ, Koelle DM, Frazer IH. Immune responses to a HSV-2 polynucleotide immunotherapy COR-1 in HSV-2 positive subjects: A randomized double blinded phase I/IIa trial. PLoS One 2019; 14:e0226320. [PMID: 31846475 PMCID: PMC6917347 DOI: 10.1371/journal.pone.0226320] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/14/2019] [Indexed: 02/03/2023] Open
Abstract
Background Genital herpes simplex infection affects more than 500 million people worldwide. We have previously shown that COR-1, a therapeutic HSV-2 polynucleotide vaccine candidate, is safe and well tolerated in healthy subjects. Objective Here, we present a single center double-blind placebo-controlled, randomized phase I/IIa trial of COR-1 in HSV-2 positive subjects in which we assessed safety and tolerability as primary endpoints, and immunogenicity and therapeutic efficacy as exploratory endpoints. Methods Forty-four HSV-2+ subjects confirmed by positive serology or pathology, and positive qPCR during baseline shedding, with a recurrent genital HSV-2 history of at least 12 months including three to nine reported lesions in 12 months prior to screening, aged 18 to 50 years females and males with given written informed consent, were randomized into two groups. Three immunizations at 4-week intervals and one booster immunization at 6 months, each of 1 mg COR-1 DNA or placebo, were administered intradermally as two injections of 500 μg each to either one forearm or both forearms. Results No serious adverse events, life-threatening events or deaths occurred throughout the study. As expected, HSV-2 infected subjects displayed gD2-specific antibody titers prior to immunization. COR-1 was associated with a reduction in viral shedding after booster administration compared with baseline. Conclusions This study confirms the previously demonstrated safety of COR-1 in humans and indicates a potential for use of COR-1 as a therapy to reduce viral shedding in HSV-2 infected subjects.
Collapse
Affiliation(s)
- Janin Chandra
- Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd), Translational Research Institute, Woolloongabba, Queensland, Australia
- University of Queensland, Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Wai-Ping Woo
- Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd), Translational Research Institute, Woolloongabba, Queensland, Australia
- University of Queensland, Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Julie L. Dutton
- Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd), Translational Research Institute, Woolloongabba, Queensland, Australia
- University of Queensland, Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Yan Xu
- Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd), Translational Research Institute, Woolloongabba, Queensland, Australia
- University of Queensland, Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Bo Li
- Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd), Translational Research Institute, Woolloongabba, Queensland, Australia
- University of Queensland, Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Sally Kinrade
- Medicines Development Limited, Southbank, Victoria, Australia
| | - Julian Druce
- Victorian Infectious Diseases Reference Laboratory, Melbourne, Victoria, Australia
- Doherty Institute, Melbourne, Victoria, Australia
| | - Neil Finlayson
- Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd), Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Paul Griffin
- Q-Pharm Pty Ltd, Brisbane, Queensland, Australia
- Department of Medicine and Infectious Diseases, Mater Hospital and Mater Medical Research Institute, Brisbane, Queensland, Australia
- The University of Queensland, Brisbane, Queensland, Australia
- QIMR Berghofer, Clinical Tropical Medicine Lab, Brisbane, Queensland, Australia
| | - Kerry J. Laing
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - David M. Koelle
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Benaroya Research Institute, Seattle, Washington, United States of America
| | - Ian H. Frazer
- Admedus Vaccines Pty Ltd (formerly Coridon Pty Ltd), Translational Research Institute, Woolloongabba, Queensland, Australia
- University of Queensland, Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
- * E-mail:
| |
Collapse
|
22
|
Increase in HSV shedding at initiation of antiretroviral therapy and decrease in shedding over time on antiretroviral therapy in HIV and HSV-2 infected persons. AIDS 2018; 32:2525-2531. [PMID: 30289810 DOI: 10.1097/qad.0000000000002002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES HIV-infected persons with chronic herpesvirus infections may experience paradoxical worsening after initiation of antiretroviral therapy (ART), but the impact of longer term ART is unclear. We evaluated the relationships between genital herpes simplex virus (HSV) shedding and ART initiation and time on therapy in HIV and HSV-2-infected persons. DESIGN Prospective observational study. METHODS Rates of HSV shedding in 45 HIV and HSV-2-infected persons on or off ART were prospectively followed over up to three, noncontiguous, 60-day periods, during which participants performed daily genital swabs for HSV detection by real-time HSV DNA PCR and reported symptoms. Initiation or discontinuation of ART was at the discretion of participants' healthcare providers. RESULTS In all, 6425 daily genital swabs were obtained from 45 persons (38 men and seven women) during 105 swabbing sessions. During the three sessions, 67, 74, and 92% of persons were on ART. HSV was detected on 26.5% of days in men and 22.3% of days in women. The overall rates of genital HSV shedding were 19.4% of days in persons not on ART, 30.2% in persons within 90 days of ART initiation, and 23.3% in persons on ART for longer than 90 days. After initiation of ART, HSV shedding decreased by 2% per month, or 23% per year (RR 0.98/month on ART; P = 0.0003 in adjusted analysis). This finding was consistent after including consideration of HIV viral load and CD4 cell count. CONCLUSIONS HSV shedding increased significantly shortly after ART initiation, but decreased with time on prolonged ART.
Collapse
|
23
|
Dong HJ, Wang ZH, Meng W, Li CC, Hu YX, Zhou L, Wang XJ. The Natural Compound Homoharringtonine Presents Broad Antiviral Activity In Vitro and In Vivo. Viruses 2018; 10:E601. [PMID: 30388805 PMCID: PMC6266276 DOI: 10.3390/v10110601] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/27/2018] [Accepted: 10/30/2018] [Indexed: 12/14/2022] Open
Abstract
To complement traditional antivirals, natural compounds that act via host targets and present high barriers to resistance are of increasing interest. In the work reported here, we detected that homoharringtonine (HHT) presents effective antiviral activity. HHT completely inhibited infections of vesicular stomatitis virus (VSV), Newcastle disease virus (NDV), and porcine epidemic diarrhea virus (PEDV) at concentrations of 50, 100, and 500 nM in cell cultures, respectively. Treatment with HHT at doses of 0.05 or 0.2 mg/kg significantly reduced viral load and relieved severe symptoms in PEDV- or NDV-infected animals. HHT treatment, however, moderately inhibited avian influenza virus (AIV) infection, suggesting its potent antiviral action is restricted to a number of classes of RNA viruses. In this study, we also observed that HHT actively inhibited herpes simplex virus type 1 (HSV-1) replication with a 50% inhibitory concentration (IC50) of 139 nM; the treatment with HHT at 1000 nM led to reductions of three orders of magnitude. Moreover, HHT antagonized the phosphorylation level of endogenous and exogenous eukaryotic initiation factor 4E (p-eIF4E), which might regulate the selective translation of specific messenger RNA (mRNA). HHT provides a starting point for further progress toward the clinical development of broad-spectrum antivirals.
Collapse
Affiliation(s)
- Hui-Jun Dong
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Zhao-Hua Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Wen Meng
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Cui-Cui Li
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Yan-Xin Hu
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Lei Zhou
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Xiao-Jia Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
24
|
Fife KH. Herpes Therapy Enters the Molecular Age. J Infect Dis 2018; 218:1689-1690. [DOI: 10.1093/infdis/jiy374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 06/28/2018] [Indexed: 11/13/2022] Open
|