24
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Predictors of Hepatitis B Cure Using Gene Therapy to Deliver DNA Cleavage Enzymes: A Mathematical Modeling Approach

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Most chronic viral infections are managed with small molecule therapies that inhibit replication but are not curative because non-replicating viral forms can persist despite decades of suppressive treatment. There are therefore numerous strategies in development to eradicate all non-replicating viruses from the body. We are currently engineering DNA cleavage enzymes that specifically target hepatitis B virus covalently closed circular DNA (HBV cccDNA), the episomal form of the virus that persists despite potent antiviral therapies. DNA cleavage enzymes, including homing endonucleases or meganucleases, zinc-finger nucleases (ZFNs), TAL effector nucleases (TALENs), and CRISPR-associated system 9 (Cas9) proteins, can disrupt specific regions of viral DNA. Because DNA repair is error prone, the virus can be neutralized after repeated cleavage events when a target sequence becomes mutated. DNA cleavage enzymes will be delivered as genes within viral vectors that enter hepatocytes. Here we develop mathematical models that describe the delivery and intracellular activity of DNA cleavage enzymes. Model simulations predict that high vector to target cell ratio, limited removal of delivery vectors by humoral immunity, and avid binding between enzyme and its DNA target will promote the highest level of cccDNA disruption. Development of de novo resistance to cleavage enzymes may occur if DNA cleavage and error prone repair does not render the viral episome replication incompetent: our model predicts that concurrent delivery of multiple enzymes which target different vital cccDNA regions, or sequential delivery of different enzymes, are both potentially useful strategies for avoiding multi-enzyme resistance. The underlying dynamics of cccDNA persistence are unlikely to impact the probability of cure provided that antiviral therapy is given concurrently during eradication trials. We conclude by describing experiments that can be used to validate the model, which will in turn provide vital information for dose selection for potential curative trials in animals and ultimately humans.

          Author Summary

          Innovative new approaches are being developed to eradicate viral infections that until recently were considered incurable. We are interested in engineering DNA cleavage enzymes that can cut and incapacitate persistent viruses. One hurdle is that these enzymes must be delivered to infected cells as genes within viral vectors that are not harmful to humans. In this paper, we developed a series of equations that describe the delivery of these enzymes to their intended targets, as well the activity of DNA cutting within the cell. While our mathematical model is catered towards hepatitis B virus infection, it is widely applicable to other infections such as HIV, as well as oncologic and metabolic diseases characterized by aberrant gene expression. Certain enzymes may bind DNA more avidly than others, while different enzymes may also bind cooperatively if targeted to different regions of viral DNA. We predict that such enzymes, if delivered efficiently to a high proportion of infected cells, will be critical to increase the probability of cure. We also demonstrate that our equations will serve as a useful tool for identifying the most important features of a curative regimen, and ultimately for guiding clinical trial dosing schedules to ensure hepatitis B eradication with the smallest number of possible doses.

          Related collections

          Most cited references45

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus

          Human hepatitis B virus (HBV) infection and HBV-related diseases remain a major public health problem. Individuals coinfected with its satellite hepatitis D virus (HDV) have more severe disease. Cellular entry of both viruses is mediated by HBV envelope proteins. The pre-S1 domain of the large envelope protein is a key determinant for receptor(s) binding. However, the identity of the receptor(s) is unknown. Here, by using near zero distance photo-cross-linking and tandem affinity purification, we revealed that the receptor-binding region of pre-S1 specifically interacts with sodium taurocholate cotransporting polypeptide (NTCP), a multiple transmembrane transporter predominantly expressed in the liver. Silencing NTCP inhibited HBV and HDV infection, while exogenous NTCP expression rendered nonsusceptible hepatocarcinoma cells susceptible to these viral infections. Moreover, replacing amino acids 157–165 of nonfunctional monkey NTCP with the human counterpart conferred its ability in supporting both viral infections. Our results demonstrate that NTCP is a functional receptor for HBV and HDV. DOI: http://dx.doi.org/10.7554/eLife.00049.001
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection.

            The capacity of HIV-1 to establish latent infection of CD4+ T cells may allow viral persistence despite immune responses and antiretroviral therapy. Measurements of infectious virus and viral RNA in plasma and of infectious virus, viral DNA and viral messenger RNA species in infected cells all suggest that HIV-1 replication continues throughout the course of infection. Uncertainty remains over what fraction of CD4+ T cells are infected and whether there are latent reservoirs for the virus. We show here that during the asymptomatic phase of infection there is an extremely low total body load of latently infected resting CD4+ T cells with replication-competent integrated provirus (<10(7) cells). The most prevalent form of HIV-1 DNA in resting and activated CD4+ T cells is a full-length, linear, unintegrated form that is not replication competent. The infection progresses even though at any given time in the lymphoid tissues integrated HIV-1 DNA is present in only a minute fraction of the susceptible populations, including resting and activated CD4+ T cells and macrophages.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Clades of Adeno-associated viruses are widely disseminated in human tissues.

              The potential for using Adeno-associated virus (AAV) as a vector for human gene therapy has stimulated interest in the Dependovirus genus. Serologic data suggest that AAV infections are prevalent in humans, although analyses of viruses and viral sequences from clinical samples are extremely limited. Molecular techniques were used in this study to successfully detect endogenous AAV sequences in 18% of all human tissues screened, with the liver and bone marrow being the most predominant sites. Sequence characterization of rescued AAV DNAs indicated a diverse array of molecular forms which segregate into clades whose members share functional and serologic similarities. One of the most predominant human clades is a hybrid of two previously described AAV serotypes, while another clade was found in humans and several species of nonhuman primates, suggesting a cross-species transmission of this virus. These data provide important information regarding the biology of parvoviruses in humans and their use as gene therapy vectors.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Comput Biol
                PLoS Comput. Biol
                plos
                ploscomp
                PLoS Computational Biology
                Public Library of Science (San Francisco, USA )
                1553-734X
                1553-7358
                July 2013
                July 2013
                4 July 2013
                : 9
                : 7
                : e1003131
                Affiliations
                [1 ]Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
                [2 ]Department of Medicine, University of Washington, Seattle, Washington, United States of America
                [3 ]Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
                John Hopkins University, United States of America
                Author notes

                The authors have declared that no competing interests exist.

                Conceived and designed the experiments: JTS KRJ. Performed the experiments: JTS DAS. Analyzed the data: JTS DAS. Contributed reagents/materials/analysis tools: JTS DAS. Wrote the paper: JTS DAS DS KRJ.

                Article
                PCOMPBIOL-D-13-00231
                10.1371/journal.pcbi.1003131
                3701691
                23861664
                4b7332d8-3621-477a-8147-af59fcc4321a
                Copyright @ 2013

                This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                : 7 February 2013
                : 22 May 2013
                Page count
                Pages: 16
                Funding
                This work was supported by NIH-supported Martin Delaney Collaboratory defeatHIV, grant U19 AI 096111, NIH K23 AI087206, and Grand Challenges Exploration Phase I grant 51763 and Phase II Grant OPP1018811 from the Bill and Melinda Gates Foundation. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article
                Biology
                Genomics
                Genomic Medicine
                Gene Therapy
                Microbiology
                Virology
                Antivirals
                Mechanisms of Resistance and Susceptibility
                Viral Persistence and Latency
                Mathematics
                Nonlinear Dynamics
                Medicine
                Infectious Diseases
                Viral Diseases
                Hepatitis
                Hepatitis B
                Infectious Disease Modeling

                Quantitative & Systems biology
                Quantitative & Systems biology

                Comments

                Comment on this article