19
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Liver-Targeting of Interferon-Alpha with Tissue-Specific Domain Antibodies

      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

          Interferon alpha (IFNα) is used for the treatment of hepatitis C infection and whilst efficacious it is associated with multiple adverse events including reduced leukocyte, erythrocyte, and platelet counts, fatigue, and depression. These events are most likely caused by systemic exposure to interferon. We therefore hypothesise that targeting the therapeutic directly to the intended site of action in the liver would reduce exposure in blood and peripheral tissue and hence improve the safety and tolerability of IFNα therapy. We genetically fused IFN to a domain antibody (dAb) specific to a hepatocyte restricted antigen, asialoglycoprotein receptor (ASGPR). Our results show that the murine IFNα2 homolog (mIFNα2) fused to an ASGPR specific dAb, termed DOM26h-196-61, could be expressed in mammalian tissue culture systems and retains the desirable biophysical properties and activity of both fusion partners when measured in vitro. Furthermore a clear increase in in vivo targeting of the liver by mIFNα2-ASGPR dAb fusion protein, compared to that observed with either unfused mIFNα2 or mIFNα2 fused to an isotype control dAb V HD2 (which does not bind ASGPR) was demonstrated using microSPECT imaging. We suggest that these findings may be applicable in the development of a liver-targeted human IFN molecule with improved safety and patient compliance in comparison to the current standard of care, which could ultimately be used as a treatment for human hepatitis virus infections.

          Related collections

          Most cited references23

          • Record: found
          • Abstract: found
          • Article: not found

          Binding activities of a repertoire of single immunoglobulin variable domains secreted from Escherichia coli.

          In antibodies, a heavy and a light chain variable domain, VH and VL, respectively, pack together and the hypervariable loops on each domain contribute to binding antigen. We find, however, that isolated VH domains with good antigen-binding affinities can also be prepared. Using the polymerase chain reaction, diverse libraries of VH genes were cloned from the spleen genomic DNA of mice immunized with either lysozyme or keyhole-limpet haemocyanin. From these libraries, VH domains were expressed and secreted from Escherichia coli. Binding activities were detected against both antigens, and two VH domains were characterized with affinities for lysozyme in the 20 nM range. Isolated variable domains may offer an alternative to monoclonal antibodies and serve as the key to building high-affinity human antibodies. We suggest the name 'single domain antibodies (dAbs)' for these antigen binding demands.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Domain antibodies: proteins for therapy.

            Occurring naturally in "heavy chain" immunoglobulins from camels, and now produced in fully human form, domain antibodies (dAbs) are the smallest known antigen-binding fragments of antibodies, ranging from 11 kDa to 15 kDa. dAbs are the robust variable regions of the heavy and light chains of immunoglobulins (VH and VL respectively). They are highly expressed in microbial cell culture, show favourable biophysical properties including solubility and temperature stability, and are well suited to selection and affinity maturation by in vitro selection systems such as phage display. dAbs are bioactive as monomers and, owing to their small size and inherent stability, can be formatted into larger molecules to create drugs with prolonged serum half-lives or other pharmacological activities.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Targeting dendritic cells with antigen-containing liposomes: a highly effective procedure for induction of antitumor immunity and for tumor immunotherapy.

              Dendritic cells (DCs) are potent stimulators of immunity, and DCs pulsed with tumor antigen ex vivo have applications in tumor immunotherapy. However, DCs are a small population of cells, and their isolation and pulsing with antigen can be impractical. Here we show that a crude preparation of plasma membrane vesicles (PMV) from the highly metastatic murine melanoma (B16-OVA) and a surrogate tumor antigen (OVA) can be targeted directly to DCs in vivo to elicit functional effects. A novel metal-chelating lipid, 3(nitrilotriacetic acid)-ditetradecylamine, was incorporated into B16-OVA-derived PMV, allowing recombinant hexahistidine-tagged forms of single chain antibody fragments to the DC surface molecules CD11c and DEC-205, to be conveniently "engrafted" onto the vesicle surface by metal-chelating linkage. The modified PMV, or similarly engrafted synthetic stealth liposomes containing OVA or OVA peptide antigen, were found to target DCs in vitro and in vivo, in experiments using flow cytometry and fluorescence confocal microscopy. When used as vaccines in syngeneic mice, the preparations stimulated strong B16-OVA-specific CTL responses in splenic T cells and a marked protection against tumor growth. Protection was dependent on the simultaneous delivery of both antigen and a DC maturation or "danger signal" signal (IFN-gamma or lipopolysaccharide). Administration of the DC-targeting vaccine to mice challenged with B16-OVA cells induced a dramatic immunotherapeutic effect and prolonged disease-free survival. The results show that the targeting of antigen to DCs in this way is highly effective at inducing immunity and protection against the tumor, with protection being at least partially dependent on the eosinophil chemokine eotaxin.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS One
                PLoS ONE
                plos
                plosone
                PLoS ONE
                Public Library of Science (San Francisco, USA )
                1932-6203
                2013
                25 February 2013
                : 8
                : 2
                : e57263
                Affiliations
                [1 ]Innovation Biopharm Discovery Unit, Biopharm R&D, GlaxoSmithKline, Cambridge, United Kingdom
                [2 ]Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
                Genentech, United States of America
                Author notes

                Competing Interests: The authors have read the journal‚s policy and have the following conflicts: All authors (except JS, JF, and JB) were employees of GlaxoSmithKline at the time this work was carried out. JS, JF, and JB are researchers at Queen Mary University of London whose work on this project was funded by GlaxoSmithKline. All relevant patents have been declared, and there are no products in development or marketed products to declare. This does not alter the authors‚ adherence to all the PLOS ONE policies on sharing data and materials, as detailed online in the guide for authors.

                Conceived and designed the experiments: EC JS MO LG AS RP MD AW. Performed the experiments: EC JS MO RP LG CM AS JF JB GD. Analyzed the data: EC JS MO LG AS RP MD AW. Contributed reagents/materials/analysis tools: EC JS MO RP LG AS. Wrote the paper: EC AW.

                [¤]

                Current address: Horizon Discovery Ltd., Cambridge, United Kingdom

                Article
                PONE-D-12-33862
                10.1371/journal.pone.0057263
                3581439
                23451195
                5bdd2566-db15-467a-9fe7-155fed7c7101
                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
                : 31 October 2012
                : 18 January 2013
                Page count
                Pages: 11
                Funding
                These authors have no support or funding to report.
                Categories
                Research Article
                Biology
                Biotechnology
                Drug Discovery
                Model Organisms
                Animal Models
                Mouse
                Medicine
                Drugs and Devices
                Pharmacokinetics
                Drug Distribution
                Drug Research and Development
                Pharmacodynamics
                Oncology
                Cancer Treatment
                Antibody Therapy
                Radiology
                Nuclear Medicine
                PET imaging
                Radionuclide Imaging
                SPECT imaging

                Uncategorized
                Uncategorized

                Comments

                Comment on this article